Aouad, P.
Quinn, H.M.
Berger, A.
Brisken, C.
(2024). Tumor dormancy: EMT beyond invasion and metastasis. Genesis,
Vol.62
(1),
p. e23552.
show abstract
More than two-thirds of cancer-related deaths are attributable to metastases. In some tumor types metastasis can occur up to 20 years after diagnosis and successful treatment of the primary tumor, a phenomenon termed late recurrence. Metastases arise from disseminated tumor cells (DTCs) that leave the primary tumor early on in tumor development, either as single cells or clusters, adapt to new environments, and reduce or shut down their proliferation entering a state of dormancy for prolonged periods of time. Dormancy has been difficult to track clinically and study experimentally. Recent advances in technology and disease modeling have provided new insights into the molecular mechanisms orchestrating dormancy and the switch to a proliferative state. A new role for epithelial-mesenchymal transition (EMT) in inducing plasticity and maintaining a dormant state in several cancer models has been revealed. In this review, we summarize the major findings linking EMT to dormancy control and highlight the importance of pre-clinical models and tumor/tissue context when designing studies. Understanding of the cellular and molecular mechanisms controlling dormant DTCs is pivotal in developing new therapeutic agents that prevent distant recurrence by maintaining a dormant state..
Ronchi, C.
Haider, S.
Brisken, C.
(2024). EMBER creates a unified space for independent breast cancer transcriptomic datasets enabling precision oncology. Npj breast cancer,
Vol.10
(1),
p. 56.
show abstract
full text
Transcriptomics has revolutionized biomedical research and refined breast cancer subtyping and diagnostics. However, wider use in clinical practice is hampered for a number of reasons including the application of transcriptomic signatures as single sample predictors. Here, we present an embedding approach called EMBER that creates a unified space of 11,000 breast cancer transcriptomes and predicts phenotypes of transcriptomic profiles on a single sample basis. EMBER accurately captures the five molecular subtypes. Key biological pathways, such as estrogen receptor signaling, cell proliferation, DNA repair, and epithelial-mesenchymal transition determine sample position in the space. We validate EMBER in four independent patient cohorts and show with samples from the window trial, POETIC, that it captures clinical responses to endocrine therapy and identifies increased androgen receptor signaling and decreased TGFβ signaling as potential mechanisms underlying intrinsic therapy resistance. Of direct clinical importance, we show that the EMBER-based estrogen receptor (ER) signaling score is superior to the immunohistochemistry (IHC) based ER index used in current clinical practice to select patients for endocrine therapy. As such, EMBER provides a calibration and reference tool that paves the way for using RNA-seq as a standard diagnostic and predictive tool for ER+ breast cancer..
Geukens, T.
De Schepper, M.
Van Den Bogaert, W.
Van Baelen, K.
Maetens, M.
Pabba, A.
Mahdami, A.
Leduc, S.
Isnaldi, E.
Nguyen, H.-.
Bachir, I.
Hajipirloo, M.
Zels, G.
Van Cauwenberge, J.
Borremans, K.
Vandecaveye, V.
Weynand, B.
Vermeulen, P.
Leucci, E.
Baietti, M.F.
Sflomos, G.
Battista, L.
Brisken, C.
Derksen, P.W.
Koorman, T.
Visser, D.
Scheele, C.L.
Thommen, D.S.
Hatse, S.
Fendt, S.-.
Vanderheyden, E.
Van Brussel, T.
Schepers, R.
Boeckx, B.
Lambrechts, D.
Marano, G.
Biganzoli, E.
Smeets, A.
Nevelsteen, I.
Punie, K.
Neven, P.
Wildiers, H.
Richard, F.
Floris, G.
Desmedt, C.
(2024). Rapid autopsies to enhance metastatic research: the UPTIDER post-mortem tissue donation program. Npj breast cancer,
Vol.10
(1),
p. 31.
show abstract
full text
Research on metastatic cancer has been hampered by limited sample availability. Here we present the breast cancer post-mortem tissue donation program UPTIDER and show how it enabled sampling of a median of 31 (range: 5-90) metastases and 5-8 liquids per patient from its first 20 patients. In a dedicated experiment, we show the mild impact of increasing time after death on RNA quality, transcriptional profiles and immunohistochemical staining in tumor tissue samples. We show that this impact can be counteracted by organ cooling. We successfully generated ex vivo models from tissue and liquid biopsies from distinct histological subtypes of breast cancer. We anticipate these and future findings of UPTIDER to elucidate mechanisms of disease progression and treatment resistance and to provide tools for the exploration of precision medicine strategies in the metastatic setting..
Flaherty, R.L.
Sflomos, G.
Brisken, C.
(2024). Is There a Special Role for Ovarian Hormones in the Pathogenesis of Lobular Carcinoma?. Endocrinology,
Vol.165
(5).
show abstract
full text
Lobular carcinoma represent the most common special histological subtype of breast cancer, with the majority classed as hormone receptor positive. Rates of invasive lobular carcinoma in postmenopausal women have been seen to increase globally, while other hormone receptor-positive breast cancers proportionally have not followed the same trend. This has been linked to exposure to exogenous ovarian hormones such as hormone replacement therapy. Reproductive factors resulting in increased lifetime exposure to endogenous ovarian hormones have also been linked to an increased risk of lobular breast cancer, and taken together, these data make a case for the role of ovarian hormones in the genesis and progression of the disease. In this review, we summarize current understanding of the epidemiological associations between ovarian hormones and lobular breast cancer and highlight mechanistic links that may underpin the etiology and biology..
Quinn, H.M.
Battista, L.
Scabia, V.
Brisken, C.
(2024). Preclinical Mouse Intraductal Model (MIND) to Study Metastatic Dormancy in Estrogen Receptor-Positive Breast Cancer. Methods mol biol,
Vol.2811,
pp. 101-112.
show abstract
Here, we describe a clinically relevant xenograft model of hormone receptor-positive breast cancer that maintains estrogen receptor (ER) status without the need for exogenous supplementation of hormones. The naturally low 17-β-estradiol levels in host mice recapitulate levels seen in post-menopausal women. By introducing breast cancer cells directly into their "natural" microenvironment of the milk ducts, these cells maintain hormone receptor status, model the clinical progression of the disease, and develop ER- metastatic lesions or dormant micrometastatic lesions in the case of ER+ BC. With the use of GFP/RFP:Luc2 reporters, we can monitor in vivo tumour growth and conduct ex vivo metastases assays to evaluate dormant metastatic cell harboring organs. Upon recovery of metastatic cells from ER+ breast cancer models, downstream analyses can be conducted to assess the relationship between epithelial plasticity and metastatic dormancy..
Ronchi, C.
Brisken, C.
(2023). Targeting the Progesterone Receptor in Breast Cancer: Mind the Short Form!. Clin cancer res,
Vol.29
(5),
pp. 833-834.
show abstract
full text
The presurgical window of opportunity trial (WOT) MIPRA provides evidence that neoadjuvant treatment with the progesterone receptor (PR) antagonist mifepristone (RU486) may benefit patients with estrogen receptor-positive (ER+) breast cancer characterized by a high ratio of PR-A versus PR-B isoform (>1.5), suggesting that PR may be targeted in a subset of patients. See related article by Elía et al., p. 866..
Gibson, S.V.
Tomas Bort, E.
Rodríguez-Fernández, L.
Allen, M.D.
Gomm, J.J.
Goulding, I.
Auf dem Keller, U.
Agnoletto, A.
Brisken, C.
Peck, B.
Cameron, A.J.
Marshall, J.F.
Jones, J.L.
Carter, E.P.
Grose, R.P.
(2023). TGFβ-mediated MMP13 secretion drives myoepithelial cell dependent breast cancer progression. Npj breast cancer,
Vol.9
(1),
p. 9.
show abstract
full text
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer. Virtually all women with DCIS are treated, despite evidence suggesting up to half would remain with stable, non-threatening, disease. Overtreatment thus presents a pressing issue in DCIS management. To understand the role of the normally tumour suppressive myoepithelial cell in disease progression we present a 3D in vitro model incorporating both luminal and myoepithelial cells in physiomimetic conditions. We demonstrate that DCIS-associated myoepithelial cells promote striking myoepithelial-led invasion of luminal cells, mediated by the collagenase MMP13 through a non-canonical TGFβ - EP300 pathway. In vivo, MMP13 expression is associated with stromal invasion in a murine model of DCIS progression and is elevated in myoepithelial cells of clinical high-grade DCIS cases. Our data identify a key role for myoepithelial-derived MMP13 in facilitating DCIS progression and point the way towards a robust marker for risk stratification in DCIS patients..
Sflomos, G.
Schaumann, N.
Christgen, M.
Christgen, H.
Bartels, S.
Kreipe, H.
Battista, L.
Brisken, C.
(2023). Optimized Modeling of Metastatic Triple-Negative Invasive Lobular Breast Carcinoma. Cancers (basel),
Vol.15
(13).
show abstract
full text
Invasive lobular carcinoma (ILC) is a common breast cancer subtype that is often diagnosed at advanced stages and causes significant morbidity. Late-onset secondary tumor recurrence affects up to 30% of ILC patients, posing a therapeutic challenge if resistance to systemic therapy develops. Nonetheless, there is a lack of preclinical models for ILC, and the current models do not accurately reproduce the complete range of the disease. We created clinically relevant metastatic xenografts to address this gap by grafting the triple-negative IPH-926 cell line into mouse milk ducts. The resulting intraductal xenografts accurately recapitulate lobular carcinoma in situ (LCIS), invasive lobular carcinoma, and metastatic ILC in relevant organs. Using a panel of 15 clinical markers, we characterized the intratumoral heterogeneity of primary and metastatic lesions. Interestingly, intraductal IPH-926 xenografts express low but actionable HER2 and are not dependent on supplementation with the ovarian hormone estradiol for their growth. This model provides a valuable tool to test the efficiency of potential new ILC therapeutics, and it may help detect vulnerabilities within ILC that can be exploited for therapeutic targeting..
Scabia, V.
Ayyanan, A.
De Martino, F.
Agnoletto, A.
Battista, L.
Laszlo, C.
Treboux, A.
Zaman, K.
Stravodimou, A.
Jallut, D.
Fiche, M.
Bucher, P.
Ambrosini, G.
Sflomos, G.
Brisken, C.
(2022). Estrogen receptor positive breast cancers have patient specific hormone sensitivities and rely on progesterone receptor. Nat commun,
Vol.13
(1),
p. 3127.
show abstract
full text
Estrogen and progesterone receptor (ER, PR) signaling control breast development and impinge on breast carcinogenesis. ER is an established driver of ER + disease but the role of the PR, itself an ER target gene, is debated. We assess the issue in clinically relevant settings by a genetic approach and inject ER + breast cancer cell lines and patient-derived tumor cells to the milk ducts of immunocompromised mice. Such ER + xenografts were exposed to physiologically relevant levels of 17-β-estradiol (E2) and progesterone (P4). We find that independently both premenopausal E2 and P4 levels increase tumor growth and combined treatment enhances metastatic spread. The proliferative responses are patient-specific with MYC and androgen receptor (AR) signatures determining P4 response. PR is required for tumor growth in patient samples and sufficient to drive tumor growth and metastasis in ER signaling ablated tumor cells. Our findings suggest that endocrine therapy may need to be personalized, and that abrogating PR expression can be a therapeutic option..
Bernier-Latmani, J.
Mauri, C.
Marcone, R.
Renevey, F.
Durot, S.
He, L.
Vanlandewijck, M.
Maclachlan, C.
Davanture, S.
Zamboni, N.
Knott, G.W.
Luther, S.A.
Betsholtz, C.
Delorenzi, M.
Brisken, C.
Petrova, T.V.
(2022). ADAMTS18+ villus tip telocytes maintain a polarized VEGFA signaling domain and fenestrations in nutrient-absorbing intestinal blood vessels. Nat commun,
Vol.13
(1),
p. 3983.
show abstract
full text
The small intestinal villus tip is the first point of contact for lumen-derived substances including nutrients and microbial products. Electron microscopy studies from the early 1970s uncovered unusual spatial organization of small intestinal villus tip blood vessels: their exterior, epithelial-facing side is fenestrated, while the side facing the villus stroma is non-fenestrated, covered by pericytes and harbors endothelial nuclei. Such organization optimizes the absorption process, however the molecular mechanisms maintaining this highly specialized structure remain unclear. Here we report that perivascular LGR5+ villus tip telocytes (VTTs) are necessary for maintenance of villus tip endothelial cell polarization and fenestration by sequestering VEGFA signaling. Mechanistically, unique VTT expression of the protease ADAMTS18 is necessary for VEGFA signaling sequestration through limiting fibronectin accumulation. Therefore, we propose a model in which LGR5+ ADAMTS18+ telocytes are necessary to maintain a "just-right" level and location of VEGFA signaling in intestinal villus blood vasculature to ensure on one hand the presence of sufficient endothelial fenestrae, while avoiding excessive leakiness of the vessels and destabilization of villus tip epithelial structures..
Staaf, J.
Häkkinen, J.
Hegardt, C.
Saal, L.H.
Kimbung, S.
Hedenfalk, I.
Lien, T.
Sørlie, T.
Naume, B.
Russnes, H.
Marcone, R.
Ayyanan, A.
Brisken, C.
Malterling, R.R.
Asking, B.
Olofsson, H.
Lindman, H.
Bendahl, P.-.
Ehinger, A.
Larsson, C.
Loman, N.
Rydén, L.
Malmberg, M.
Borg, Å.
Vallon-Christersson, J.
(2022). RNA sequencing-based single sample predictors of molecular subtype and risk of recurrence for clinical assessment of early-stage breast cancer. Npj breast cancer,
Vol.8
(1),
p. 94.
show abstract
full text
Multigene assays for molecular subtypes and biomarkers can aid management of early invasive breast cancer. Using RNA-sequencing we aimed to develop single-sample predictor (SSP) models for clinical markers, subtypes, and risk of recurrence (ROR). A cohort of 7743 patients was divided into training and test set. We trained SSPs for subtypes and ROR assigned by nearest-centroid (NC) methods and SSPs for biomarkers from histopathology. Classifications were compared with Prosigna in two external cohorts (ABiM, n = 100 and OSLO2-EMIT0, n = 103). Prognostic value was assessed using distant recurrence-free interval. Agreement between SSP and NC for PAM50 (five subtypes) was high (85%, Kappa = 0.78) for Subtype (four subtypes) very high (90%, Kappa = 0.84) and for ROR risk category high (84%, Kappa = 0.75, weighted Kappa = 0.90). Prognostic value was assessed as equivalent and clinically relevant. Agreement with histopathology was very high or high for receptor status, while moderate for Ki67 status and poor for Nottingham histological grade. SSP and Prosigna concordance was high for subtype (OSLO-EMIT0 83%, Kappa = 0.73 and ABiM 80%, Kappa = 0.72) and moderate and high for ROR risk category (68 and 84%, Kappa = 0.50 and 0.70, weighted Kappa = 0.70 and 0.78). Pooled concordance for emulated treatment recommendation dichotomized for chemotherapy was high (85%, Kappa = 0.66). Retrospective evaluation suggested that SSP application could change chemotherapy recommendations for up to 17% of postmenopausal ER+/HER2-/N0 patients with balanced escalation and de-escalation. Results suggest that NC and SSP models are interchangeable on a group-level and nearly so on a patient level and that SSP models can be derived to closely match clinical tests..
Aouad, P.
Zhang, Y.
De Martino, F.
Stibolt, C.
Ali, S.
Ambrosini, G.
Mani, S.A.
Maggs, K.
Quinn, H.M.
Sflomos, G.
Brisken, C.
(2022). Epithelial-mesenchymal plasticity determines estrogen receptor positive breast cancer dormancy and epithelial reconversion drives recurrence. Nat commun,
Vol.13
(1),
p. 4975.
show abstract
full text
More than 70% of human breast cancers (BCs) are estrogen receptor α-positive (ER+). A clinical challenge of ER+ BC is that they can recur decades after initial treatments. Mechanisms governing latent disease remain elusive due to lack of adequate in vivo models. We compare intraductal xenografts of ER+ and triple-negative (TN) BC cells and demonstrate that disseminated TNBC cells proliferate similarly as TNBC cells at the primary site whereas disseminated ER+ BC cells proliferate slower, they decrease CDH1 and increase ZEB1,2 expressions, and exhibit characteristics of epithelial-mesenchymal plasticity (EMP) and dormancy. Forced E-cadherin expression overcomes ER+ BC dormancy. Cytokine signalings are enriched in more active versus inactive disseminated tumour cells, suggesting microenvironmental triggers for awakening. We conclude that intraductal xenografts model ER + BC dormancy and reveal that EMP is essential for the generation of a dormant cell state and that targeting exit from EMP has therapeutic potential..
Sflomos, G.
Battista, L.
Aouad, P.
De Martino, F.
Scabia, V.
Stravodimou, A.
Ayyanan, A.
Ifticene-Treboux, A.
RLS,
Bucher, P.
Fiche, M.
Ambrosini, G.
Brisken, C.
(2021). Intraductal xenografts show lobular carcinoma cells rely on their own extracellular matrix and LOXL1. Embo mol med,
Vol.13
(3),
p. e13180.
show abstract
full text
Invasive lobular carcinoma (ILC) is the most frequent special histological subtype of breast cancer, typically characterized by loss of E-cadherin. It has clinical features distinct from other estrogen receptor-positive (ER+ ) breast cancers but the molecular mechanisms underlying its characteristic biology are poorly understood because we lack experimental models to study them. Here, we recapitulate the human disease, including its metastatic pattern, by grafting ILC-derived breast cancer cell lines, SUM-44 PE and MDA-MB-134-VI cells, into the mouse milk ducts. Using patient-derived intraductal xenografts from lobular and non-lobular ER+ HER2- tumors to compare global gene expression, we identify extracellular matrix modulation as a lobular carcinoma cell-intrinsic trait. Analysis of TCGA patient datasets shows matrisome signature is enriched in lobular carcinomas with overexpression of elastin, collagens, and the collagen modifying enzyme LOXL1. Treatment with the pan LOX inhibitor BAPN and silencing of LOXL1 expression decrease tumor growth, invasion, and metastasis by disrupting ECM structure resulting in decreased ER signaling. We conclude that LOXL1 inhibition is a promising therapeutic strategy for ILC..
Juppet, Q.
De Martino, F.
Marcandalli, E.
Weigert, M.
Burri, O.
Unser, M.
Brisken, C.
Sage, D.
(2021). Deep Learning Enables Individual Xenograft Cell Classification in Histological Images by Analysis of Contextual Features. J mammary gland biol neoplasia,
Vol.26
(2),
pp. 101-112.
show abstract
full text
Patient-Derived Xenografts (PDXs) are the preclinical models which best recapitulate inter- and intra-patient complexity of human breast malignancies, and are also emerging as useful tools to study the normal breast epithelium. However, data analysis generated with such models is often confounded by the presence of host cells and can give rise to data misinterpretation. For instance, it is important to discriminate between xenografted and host cells in histological sections prior to performing immunostainings. We developed Single Cell Classifier (SCC), a data-driven deep learning-based computational tool that provides an innovative approach for automated cell species discrimination based on a multi-step process entailing nuclei segmentation and single cell classification. We show that human and murine cell contextual features, more than cell-intrinsic ones, can be exploited to discriminate between cell species in both normal and malignant tissues, yielding up to 96% classification accuracy. SCC will facilitate the interpretation of H&E- and DAPI-stained histological sections of xenografted human-in-mouse tissues and it is open to new in-house built models for further applications. SCC is released as an open-source plugin in ImageJ/Fiji available at the following link: https://github.com/Biomedical-Imaging-Group/SingleCellClassifier ..
Shamseddin, M.
De Martino, F.
Constantin, C.
Scabia, V.
Lancelot, A.-.
Laszlo, C.
Ayyannan, A.
Battista, L.
Raffoul, W.
Gailloud-Matthieu, M.-.
Bucher, P.
Fiche, M.
Ambrosini, G.
Sflomos, G.
Brisken, C.
(2021). Contraceptive progestins with androgenic properties stimulate breast epithelial cell proliferation. Embo mol med,
Vol.13
(7),
p. e14314.
show abstract
full text
Hormonal contraception exposes women to synthetic progesterone receptor (PR) agonists, progestins, and transiently increases breast cancer risk. How progesterone and progestins affect the breast epithelium is poorly understood because we lack adequate models to study this. We hypothesized that individual progestins differentially affect breast epithelial cell proliferation and hence breast cancer risk. Using mouse mammary tissue ex vivo, we show that testosterone-related progestins induce the PR target and mediator of PR signaling-induced cell proliferation receptor activator of NF-κB ligand (Rankl), whereas progestins with anti-androgenic properties in reporter assays do not. We develop intraductal xenografts of human breast epithelial cells from 36 women, show they remain hormone-responsive and that progesterone and the androgenic progestins, desogestrel, gestodene, and levonorgestrel, promote proliferation but the anti-androgenic, chlormadinone, and cyproterone acetate, do not. Prolonged exposure to androgenic progestins elicits hyperproliferation with cytologic changes. Androgen receptor inhibition interferes with PR agonist- and levonorgestrel-induced RANKL expression and reduces levonorgestrel-driven cell proliferation. Thus, different progestins have distinct biological activities in the breast epithelium to be considered for more informed choices in hormonal contraception..
Sflomos, G.
Schipper, K.
Koorman, T.
Fitzpatrick, A.
Oesterreich, S.
Lee, A.V.
Jonkers, J.
Brunton, V.G.
Christgen, M.
Isacke, C.
Derksen, P.W.
Brisken, C.
(2021). Atlas of Lobular Breast Cancer Models: Challenges and Strategic Directions. Cancers (basel),
Vol.13
(21).
show abstract
full text
Invasive lobular carcinoma (ILC) accounts for up to 15% of all breast cancer (BC) cases and responds well to endocrine treatment when estrogen receptor α-positive (ER+) yet differs in many biological aspects from other ER+ BC subtypes. Up to 30% of patients with ILC will develop late-onset metastatic disease up to ten years after initial tumor diagnosis and may experience failure of systemic therapy. Unfortunately, preclinical models to study ILC progression and predict the efficacy of novel therapeutics are scarce. Here, we review the current advances in ILC modeling, including cell lines and organotypic models, genetically engineered mouse models, and patient-derived xenografts. We also underscore four critical challenges that can be addressed using ILC models: drug resistance, lobular tumor microenvironment, tumor dormancy, and metastasis. Finally, we highlight the advantages of shared experimental ILC resources and provide essential considerations from the perspective of the European Lobular Breast Cancer Consortium (ELBCC), which is devoted to better understanding and translating the molecular cues that underpin ILC to clinical diagnosis and intervention. This review will guide investigators who are considering the implementation of ILC models in their research programs..
Koch, C.
Kuske, A.
Joosse, S.A.
Yigit, G.
Sflomos, G.
Thaler, S.
Smit, D.J.
Werner, S.
Borgmann, K.
Gärtner, S.
Mossahebi Mohammadi, P.
Battista, L.
Cayrefourcq, L.
Altmüller, J.
Salinas-Riester, G.
Raithatha, K.
Zibat, A.
Goy, Y.
Ott, L.
Bartkowiak, K.
Tan, T.Z.
Zhou, Q.
Speicher, M.R.
Müller, V.
Gorges, T.M.
Jücker, M.
Thiery, J.-.
Brisken, C.
Riethdorf, S.
Alix-Panabières, C.
Pantel, K.
(2020). Characterization of circulating breast cancer cells with tumorigenic and metastatic capacity. Embo mol med,
Vol.12
(9),
p. e11908.
show abstract
full text
Functional studies giving insight into the biology of circulating tumor cells (CTCs) remain scarce due to the low frequency of CTCs and lack of appropriate models. Here, we describe the characterization of a novel CTC-derived breast cancer cell line, designated CTC-ITB-01, established from a patient with metastatic estrogen receptor-positive (ER+ ) breast cancer, resistant to endocrine therapy. CTC-ITB-01 remained ER+ in culture, and copy number alteration (CNA) profiling showed high concordance between CTC-ITB-01 and CTCs originally present in the patient with cancer at the time point of blood draw. RNA-sequencing data indicate that CTC-ITB-01 has a predominantly epithelial expression signature. Primary tumor and metastasis formation in an intraductal PDX mouse model mirrored the clinical progression of ER+ breast cancer. Downstream ER signaling was constitutively active in CTC-ITB-01 independent of ligand availability, and the CDK4/6 inhibitor Palbociclib strongly inhibited CTC-ITB-01 growth. Thus, we established a functional model that opens a new avenue to study CTC biology..
Brisken, C.
Scabia, V.
(2020). 90 YEARS OF PROGESTERONE: Progesterone receptor signaling in the normal breast and its implications for cancer. J mol endocrinol,
Vol.65
(1),
pp. T81-T94.
show abstract
full text
Progesterone is considered as the pregnancy hormone and acts on many different target tissues. Progesterone receptor (PR) signaling is important for normal development and the physiologic function of the breast and impinges on breast carcinogenesis. Both systemically and locally, in the breast epithelium, there are multiple layers of complexity to progesterone action, many of which have been revealed through experiments in mice. The hormone acts via its receptor expressed in a subset of cells, the sensor cells, in the breast epithelium with different signaling outcomes in individual cells eliciting distinct cell-intrinsic and paracrine signaling involving different mediators for different intercellular interactions. PR expression itself is developmentally regulated and the biological outcome of PR signaling depends on the developmental stage of the mammary gland and the endocrine context. During both puberty and adulthood PR activates stem and progenitor cells through Wnt4-driven activation of the myoepithelium with downstream Adamts18-induced changes in extracellualr matrix (ECM) / basal membrane (BM). During estrous cycling and pregnancy, the hormone drives a major cell expansion through Rankl. At all stages, PR signaling is closely tied to estrogen receptor α (ER) signaling. As the PR itself is a target gene of ER, the complex interactions are experimentally difficult to dissect and still poorly understood. Ex vivo models of the human breast and studies on biopsy samples show that major signaling axes are conserved across species. New intraductal xenograft models hold promise to provide a better understanding of PR signaling in the normal breast epithelium and in breast cancer development in the near future..
Gagniac, L.
Rusidzé, M.
Boudou, F.
Cagnet, S.
Adlanmerini, M.
Jeannot, P.
Gaide, N.
Giton, F.
Besson, A.
Weyl, A.
Gourdy, P.
Raymond-Letron, I.
Arnal, J.-.
Brisken, C.
Lenfant, F.
(2020). Membrane expression of the estrogen receptor ERα is required for intercellular communications in the mammary epithelium. Development,
Vol.147
(5).
show abstract
full text
17β-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development..
Siersbæk, R.
Scabia, V.
Nagarajan, S.
Chernukhin, I.
Papachristou, E.K.
Broome, R.
Johnston, S.J.
Joosten, S.E.
Green, A.R.
Kumar, S.
Jones, J.
Omarjee, S.
Alvarez-Fernandez, R.
Glont, S.
Aitken, S.J.
Kishore, K.
Cheeseman, D.
Rakha, E.A.
D'Santos, C.
Zwart, W.
Russell, A.
Brisken, C.
Carroll, J.S.
(2020). IL6/STAT3 Signaling Hijacks Estrogen Receptor α Enhancers to Drive Breast Cancer Metastasis. Cancer cell,
Vol.38
(3),
pp. 412-423.e9.
show abstract
full text
The cytokine interleukin-6 (IL6) and its downstream effector STAT3 constitute a key oncogenic pathway, which has been thought to be functionally connected to estrogen receptor α (ER) in breast cancer. We demonstrate that IL6/STAT3 signaling drives metastasis in ER+ breast cancer independent of ER. STAT3 hijacks a subset of ER enhancers to drive a distinct transcriptional program. Although these enhancers are shared by both STAT3 and ER, IL6/STAT3 activity is refractory to standard ER-targeted therapies. Instead, inhibition of STAT3 activity using the JAK inhibitor ruxolitinib decreases breast cancer invasion in vivo. Therefore, IL6/STAT3 and ER oncogenic pathways are functionally decoupled, highlighting the potential of IL6/STAT3-targeted therapies in ER+ breast cancer..
Cartaxo, A.L.
Estrada, M.F.
Domenici, G.
Roque, R.
Silva, F.
Gualda, E.J.
Loza-Alvarez, P.
Sflomos, G.
Brisken, C.
Alves, P.M.
André, S.
Brito, C.
(2020). A novel culture method that sustains ERα signaling in human breast cancer tissue microstructures. J exp clin cancer res,
Vol.39
(1),
p. 161.
show abstract
full text
BACKGROUND: Estrogen receptor α (ERα) signaling is a defining and driving event in most breast cancers; ERα is detected in malignant epithelial cells of 75% of all breast cancers (classified as ER-positive breast cancer) and, in these cases, ERα targeting is the main therapeutic strategy. However, the biological determinants of ERα heterogeneity and the mechanisms underlying therapeutic resistance are still elusive, hampered by the challenges in developing experimental models recapitulative of intra-tumoral heterogeneity and in which ERα signaling is sustained. Ex vivo cultures of human breast cancer tissue have been proposed to retain the original tissue architecture, epithelial and stromal cell components and ERα. However, loss of cellularity, viability and ERα expression are well-known culture-related phenomena. METHODS: BC samples were collected and brought to the laboratory. Then they were minced, enzymatically digested, entrapped in alginate and cultured for 1 month. The histological architecture, cellular composition and cell proliferation of tissue microstructures were assessed by immunohistochemistry. Cell viability was assessed by measurement of cell metabolic activity and histological evaluation. The presence of ERα was accessed by immunohistochemistry and RT-qPCR and its functionality evaluated by challenge with 17-β-estradiol and fulvestrant. RESULTS: We describe a strategy based on entrapment of breast cancer tissue microstructures in alginate capsules and their long-term culture under agitation, successfully applied to tissue obtained from 63 breast cancer patients. After 1 month in culture, the architectural features of the encapsulated tissue microstructures were similar to the original patient tumors: epithelial, stromal and endothelial compartments were maintained, with an average of 97% of cell viability compared to day 0. In ERα-positive cases, fibers of collagen, the main extracellular matrix component in vivo, were preserved. ERα expression was at least partially retained at gene and protein levels and response to ERα stimulation and inhibition was observed at the level of downstream targets, demonstrating active ER signaling. CONCLUSIONS: The proposed model system is a new methodology to study ex vivo breast cancer biology, in particular ERα signaling. It is suitable for interrogating the long-term effects of anti-endocrine drugs in a set-up that closely resembles the original tumor microenvironment, with potential application in pre- and co-clinical assays of ERα-positive breast cancer..
Ataca, D.
Aouad, P.
Constantin, C.
Laszlo, C.
Beleut, M.
Shamseddin, M.
Rajaram, R.D.
Jeitziner, R.
Mead, T.J.
Caikovski, M.
Bucher, P.
Ambrosini, G.
Apte, S.S.
Brisken, C.
(2020). The secreted protease Adamts18 links hormone action to activation of the mammary stem cell niche. Nat commun,
Vol.11
(1),
p. 1571.
show abstract
full text
Estrogens and progesterone control breast development and carcinogenesis via their cognate receptors expressed in a subset of luminal cells in the mammary epithelium. How they control the extracellular matrix, important to breast physiology and tumorigenesis, remains unclear. Here we report that both hormones induce the secreted protease Adamts18 in myoepithelial cells by controlling Wnt4 expression with consequent paracrine canonical Wnt signaling activation. Adamts18 is required for stem cell activation, has multiple binding partners in the basement membrane and interacts genetically with the basal membrane-specific proteoglycan, Col18a1, pointing to the basement membrane as part of the stem cell niche. In vitro, ADAMTS18 cleaves fibronectin; in vivo, Adamts18 deletion causes increased collagen deposition during puberty, which results in impaired Hippo signaling and reduced Fgfr2 expression both of which control stem cell function. Thus, Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation..
Fiche, M.
Scabia, V.
Aouad, P.
Battista, L.
Treboux, A.
Stravodimou, A.
Zaman, K.
RLS,
Dormoy, V.
Ayyanan, A.
Sflomos, G.
Brisken, C.
(2019). Intraductal patient-derived xenografts of estrogen receptor α-positive breast cancer recapitulate the histopathological spectrum and metastatic potential of human lesions. J pathol,
Vol.247
(3),
pp. 287-292.
show abstract
full text
Estrogen receptor α-positive (ER-positive) or 'luminal' breast cancers were notoriously difficult to establish as patient-derived xenografts (PDXs). We and others recently demonstrated that the microenvironment is critical for ER-positive tumor cells; when grafted as single cells into milk ducts of NOD Scid gamma females, >90% of ER-positive tumors can be established as xenografts and recapitulate many features of the human disease in vivo. This intraductal approach holds promise for personalized medicine, yet human and murine stroma are organized differently and this and other species specificities may limit the value of this model. Here, we analyzed 21 ER-positive intraductal PDXs histopathologically. We found that intraductal PDXs vary in extent and define four histopathological patterns: flat, lobular, in situ and invasive, which occur in pure and combined forms. The intraductal PDXs replicate earlier stages of tumor development than their clinical counterparts. Micrometastases are already detected when lesions appear in situ. Tumor extent, histopathological patterns and micrometastatic load correlate with biological properties of their tumors of origin. Our findings add evidence to the validity of the intraductal model for in vivo studies of ER-positive breast cancer and raise the intriguing possibility that tumor cell dissemination may occur earlier than currently thought. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland..
Jeitziner, R.
Carrière, M.
Rougemont, J.
Oudot, S.
Hess, K.
Brisken, C.
(2019). Two-Tier Mapper, an unbiased topology-based clustering method for enhanced global gene expression analysis. Bioinformatics,
Vol.35
(18),
pp. 3339-3347.
show abstract
MOTIVATION: Unbiased clustering methods are needed to analyze growing numbers of complex datasets. Currently available clustering methods often depend on parameters that are set by the user, they lack stability, and are not applicable to small datasets. To overcome these shortcomings we used topological data analysis, an emerging field of mathematics that discerns additional feature and discovers hidden insights on datasets and has a wide application range. RESULTS: We have developed a topology-based clustering method called Two-Tier Mapper (TTMap) for enhanced analysis of global gene expression datasets. First, TTMap discerns divergent features in the control group, adjusts for them, and identifies outliers. Second, the deviation of each test sample from the control group in a high-dimensional space is computed, and the test samples are clustered using a new Mapper-based topological algorithm at two levels: a global tier and local tiers. All parameters are either carefully chosen or data-driven, avoiding any user-induced bias. The method is stable, different datasets can be combined for analysis, and significant subgroups can be identified. It outperforms current clustering methods in sensitivity and stability on synthetic and biological datasets, in particular when sample sizes are small; outcome is not affected by removal of control samples, by choice of normalization, or by subselection of data. TTMap is readily applicable to complex, highly variable biological samples and holds promise for personalized medicine. AVAILABILITY AND IMPLEMENTATION: TTMap is supplied as an R package in Bioconductor. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online..
Nacht, A.S.
Ferrari, R.
Zaurin, R.
Scabia, V.
Carbonell-Caballero, J.
Le Dily, F.
Quilez, J.
Leopoldi, A.
Brisken, C.
Beato, M.
Vicent, G.P.
(2019). C/EBPα mediates the growth inhibitory effect of progestins on breast cancer cells. Embo j,
Vol.38
(18),
p. e101426.
show abstract
full text
Steroid hormones are key gene regulators in breast cancer cells. While estrogens stimulate cell proliferation, progestins activate a single cell cycle followed by proliferation arrest. Here, we use biochemical and genome-wide approaches to show that progestins achieve this effect via a functional crosstalk with C/EBPα. Using ChIP-seq, we identify around 1,000 sites where C/EBPα binding precedes and helps binding of progesterone receptor (PR) in response to hormone. These regions exhibit epigenetic marks of active enhancers, and C/EBPα maintains an open chromatin conformation that facilitates loading of ligand-activated PR. Prior to hormone exposure, C/EBPα favors promoter-enhancer contacts that assure hormonal regulation of key genes involved in cell proliferation by facilitating binding of RAD21, YY1, and the Mediator complex. Knockdown of C/EBPα disrupts enhancer-promoter contacts and decreases the presence of these architectural proteins, highlighting its key role in 3D chromatin looping. Thus, C/EBPα fulfills a previously unknown function as a potential growth modulator in hormone-dependent breast cancer..
Laszlo, C.F.
Paz Montoya, J.
Shamseddin, M.
De Martino, F.
Beguin, A.
Nellen, R.
Bruce, S.J.
Moniatte, M.
Henry, H.
Brisken, C.
(2019). A high resolution LC-MS targeted method for the concomitant analysis of 11 contraceptive progestins and 4 steroids. J pharm biomed anal,
Vol.175,
p. 112756.
show abstract
full text
In the context of hormonal contraception and hormone replacement therapy (HRT), many women are exposed to exogenous hormones. Current use of hormonal contraception with combined ethinyl estradiol and different progestins bestows a breast cancer relative risk (RR) of 1.2- while combined HRT has a RR of 2. Although these exposures present an important public health issue, little is known about the effects of individual progestins on the breast and other tissues. Increasing availability of large scale biobanks, high throughput analyses and data management tools enable ever expanding, sophisticated population studies. In order to address the impact of distinct progestins on various health indicators, it is desirable to accurately quantify progestins in clinical samples. Here we have developed and validated a high resolution liquid chromatography mass spectrometry (LC-MS) targeted method for the simultaneous quantification of 11 synthetic progestins widely used in oral contraceptives, gestodene, levonorgestrel, etonogestrel, chlormadinone acetate, cyproterone acetate, drospirenone, desacetyl norgestimate, medroxyprogesterone acetate, norethindrone, dienogest, nomegestrol acetate, and 4 endogenous steroid hormones, progesterone, testosterone, androstenedione, and cortisol in blood samples. This highly specific quantitative analysis with high resolution Orbitrap technology detects and quantifies 15 compounds using their internal standard counterparts in a single 12 min LC-MS run. Sensitivity is attained by the use of the instrument in targeted selected ion monitoring mode. Lower limit of quantitation ranges from 2.4 pg/ml for drospirenone to 78.1 pg/ml for chlormadinone acetate. The method provides comprehensive progestin panel measurements with as little as 50 μl of murine or human plasma..
Özdemir, B.C.
Sflomos, G.
Brisken, C.
(2018). The challenges of modeling hormone receptor-positive breast cancer in mice. Endocr relat cancer,
Vol.25
(5),
pp. R319-R330.
show abstract
Estrogen receptor-positive (ER+) tumors account for 70-80% of all breast cancer (BC) cases and are characterized by estrogen dependency for their growth. Endocrine therapies using estrogen receptor antagonists or aromatase inhibitors represent a key component of the standard of care for these tumors. The occurrence of de novo or acquired resistance to estrogen withdrawal represents an important clinical problem, impacting on patient survival. In addition, despite an initially favorable outcome, a part of ER+ BC patients present with disease recurrence locally or at distant sites years or even decades after apparent remission. In vivo models that closely mimic human disease are urgently needed to study the biology of these tumors, investigate the molecular mechanisms underlying endocrine resistance and identify patients at risk of recurrence. Despite the similarities in the overall hormonal regulation of mammary gland development between mice and humans, the majority of the mammary carcinomas occurring in genetically engineered mouse models (GEMMs) are ER negative and most xenograft models are based on few ER+ cancer cell lines. We recently showed that the microenvironment is critical for ER+ cancer cells and discuss in this review the potential of intraductal xenograft model for basic and preclinical research..
Cagnet, S.
Ataca, D.
Sflomos, G.
Aouad, P.
Schuepbach-Mallepell, S.
Hugues, H.
Krust, A.
Ayyanan, A.
Scabia, V.
Brisken, C.
(2018). Oestrogen receptor α AF-1 and AF-2 domains have cell population-specific functions in the mammary epithelium. Nat commun,
Vol.9
(1),
p. 4723.
show abstract
full text
Oestrogen receptor α (ERα) is a transcription factor with ligand-independent and ligand-dependent activation functions (AF)-1 and -2. Oestrogens control postnatal mammary gland development acting on a subset of mammary epithelial cells (MECs), termed sensor cells, which are ERα-positive by immunohistochemistry (IHC) and secrete paracrine factors, which stimulate ERα-negative responder cells. Here we show that deletion of AF-1 or AF-2 blocks pubertal ductal growth and subsequent development because both are required for expression of essential paracrine mediators. Thirty percent of the luminal cells are ERα-negative by IHC but express Esr1 transcripts. This low level ERα expression through AF-2 is essential for cell expansion during puberty and growth-inhibitory during pregnancy. Cell-intrinsic ERα is not required for cell proliferation nor for secretory differentiation but controls transcript levels of cell motility and cell adhesion genes and a stem cell and epithelial mesenchymal transition (EMT) signature identifying ERα as a key regulator of mammary epithelial cell plasticity..
George, A.L.
Boulanger, C.A.
Anderson, L.H.
Cagnet, S.
Brisken, C.
Smith, G.H.
(2017). In vivo reprogramming of non-mammary cells to an epithelial cell fate is independent of amphiregulin signaling. J cell sci,
Vol.130
(12),
pp. 2018-2025.
show abstract
full text
Amphiregulin (AREG)-/- mice demonstrate impaired mammary development and form only rudimentary ductal epithelial trees; however, AREG-/- glands are still capable of undergoing alveologenesis and lactogenesis during pregnancy. Transplantation of AREG-/- mammary epithelial cells into cleared mouse mammary fat pads results in a diminished capacity for epithelial growth (∼15%) as compared to that of wild-type mammary epithelial cells. To determine whether estrogen receptor α (ERα, also known as ESR1) and/or AREG signaling were necessary for non-mammary cell redirection, we inoculated either ERα-/- or AREG-/- mammary cells with non-mammary progenitor cells (WAP-Cre/Rosa26LacZ+ male testicular cells or GFP-positive embryonic neuronal stem cells). ERα-/- cells possessed a limited ability to grow or reprogram non-mammary cells in transplanted mammary fat pads. AREG-/- mammary cells were capable of redirecting both types of non-mammary cell populations to mammary phenotypes in regenerating mammary outgrowths. Transplantation of fragments from AREG-reprogrammed chimeric outgrowths resulted in secondary outgrowths in six out of ten fat pads, demonstrating the self-renewing capacity of the redirected non-mammary cells to contribute new progeny to chimeric outgrowths. Nestin was detected at the leading edges of developing alveoli, suggesting that its expression may be essential for lobular expansion..
Dobrolecki, L.E.
Airhart, S.D.
Alferez, D.G.
Aparicio, S.
Behbod, F.
Bentires-Alj, M.
Brisken, C.
Bult, C.J.
Cai, S.
Clarke, R.B.
Dowst, H.
Ellis, M.J.
Gonzalez-Suarez, E.
Iggo, R.D.
Kabos, P.
Li, S.
Lindeman, G.J.
Marangoni, E.
McCoy, A.
Meric-Bernstam, F.
Piwnica-Worms, H.
Poupon, M.-.
Reis-Filho, J.
Sartorius, C.A.
Scabia, V.
Sflomos, G.
Tu, Y.
Vaillant, F.
Visvader, J.E.
Welm, A.
Wicha, M.S.
Lewis, M.T.
(2016). Patient-derived xenograft (PDX) models in basic and translational breast cancer research. Cancer metastasis rev,
Vol.35
(4),
pp. 547-573.
show abstract
full text
Patient-derived xenograft (PDX) models of a growing spectrum of cancers are rapidly supplanting long-established traditional cell lines as preferred models for conducting basic and translational preclinical research. In breast cancer, to complement the now curated collection of approximately 45 long-established human breast cancer cell lines, a newly formed consortium of academic laboratories, currently from Europe, Australia, and North America, herein summarizes data on over 500 stably transplantable PDX models representing all three clinical subtypes of breast cancer (ER+, HER2+, and "Triple-negative" (TNBC)). Many of these models are well-characterized with respect to genomic, transcriptomic, and proteomic features, metastatic behavior, and treatment response to a variety of standard-of-care and experimental therapeutics. These stably transplantable PDX lines are generally available for dissemination to laboratories conducting translational research, and contact information for each collection is provided. This review summarizes current experiences related to PDX generation across participating groups, efforts to develop data standards for annotation and dissemination of patient clinical information that does not compromise patient privacy, efforts to develop complementary data standards for annotation of PDX characteristics and biology, and progress toward "credentialing" of PDX models as surrogates to represent individual patients for use in preclinical and co-clinical translational research. In addition, this review highlights important unresolved questions, as well as current limitations, that have hampered more efficient generation of PDX lines and more rapid adoption of PDX use in translational breast cancer research..
Sflomos, G.
Dormoy, V.
Metsalu, T.
Jeitziner, R.
Battista, L.
Scabia, V.
Raffoul, W.
Delaloye, J.-.
Treboux, A.
Fiche, M.
Vilo, J.
Ayyanan, A.
Brisken, C.
(2016). A Preclinical Model for ERα-Positive Breast Cancer Points to the Epithelial Microenvironment as Determinant of Luminal Phenotype and Hormone Response. Cancer cell,
Vol.29
(3),
pp. 407-422.
show abstract
Seventy-five percent of breast cancers are estrogen receptor α positive (ER⁺). Research on these tumors is hampered by lack of adequate in vivo models; cell line xenografts require non-physiological hormone supplements, and patient-derived xenografts (PDXs) are hard to establish. We show that the traditional grafting of ER⁺ tumor cells into mammary fat pads induces TGFβ/SLUG signaling and basal differentiation when they require low SLUG levels to grow in vivo. Grafting into the milk ducts suppresses SLUG; ER⁺ tumor cells develop, like their clinical counterparts, in the presence of physiological hormone levels. Intraductal ER⁺ PDXs are retransplantable, predictive, and appear genomically stable. The model provides opportunities for translational research and the study of physiologically relevant hormone action in breast carcinogenesis..
Ataca, D.
Caikovski, M.
Piersigilli, A.
Moulin, A.
Benarafa, C.
Earp, S.E.
Guri, Y.
Kostic, C.
Arsenivic, Y.
Soininen, R.
Apte, S.S.
Brisken, C.
(2016). Adamts18 deletion results in distinct developmental defects and provides a model for congenital disorders of lens, lung, and female reproductive tract development. Biology open,
Vol.5,
pp. 1585-1594.
show abstract
full text
The ADAMTS family comprises 19 secreted metalloproteinases that cleave extracellular matrix components and have diverse functions in numerous disease and physiological contexts. A number of them remain ‘orphan’ proteases and among them is ADAMTS18, which has been implicated in developmental eye disorders, platelet function and various malignancies. To assess in vivo function of ADAMTS18, we generated a mouse strain with inactivated Adamts18 alleles. In the C57Bl6/Ola background, Adamts18-deficient mice are born in a normal Mendelian ratio, and are viable but show a transient growth delay. Histological examination revealed a 100% penetrant eye defect resulting from leakage of lens material through the lens capsule occurring at embryonic day (E) 13.5, when the lens grows rapidly. Adamts18-deficient lungs showed altered bronchiolar branching. Fifty percent of mutant females are infertile because of vaginal obstruction due to either a dorsoventral vaginal septum or imperforate vagina. The incidence of ovarian rete is increased in the mutant mouse strain. Thus, Adamts18 is essential in the development of distinct tissues and the new mouse strain is likely to be useful for investigating ADAMTS18 function in human disease, particularly in the contexts of infertility and carcinogenesis..
Blom, S.
Wang, Y.
Metsalu, T.
Vesterinen, T.
Pellinen, T.
Grote, A.
Linder, N.
Säilä, J.
Välimäki, K.
Kovanen, R.-.
Monni, O.
Kovanen, P.
Davies, E.
Stock, K.
Estrada, M.
Sflomos, G.
Grünewald, S.
Brito, C.
Schüler, J.
de Hoogt, R.
Brisken, C.
van der Kuip, H.
van Weerden, W.
Barry, S.
Sommergruber, W.
Anderson, E.
Nees, M.
Klefström, J.
Vilo, J.
Verschuren, E.
Graeser, R.
Hickman, J.
Lundin, J.
Kallioniemi, O.
(2015). Abstract 1698: Systems pathology for characterization of cancer model systems in a multicenter IMI-PREDECT project. Cancer research,
Vol.75
(15_Supplement),
pp. 1698-1698.
show abstract
Abstract
Despite of our increased understanding of cancer biology, the majority of anti-cancer therapies fail at late-stage clinical trials. Thus, there is an urgent need to develop and validate novel preclinical models that could predict drug efficacy in humans. For this purpose, as a part of IMI-PREDECT public-private research consortium, this study describes methodology and infrastructure for characterization and comparison of preclinical models for drug target validation applying a systems pathology approach. Formalin-fixed paraffin-embedded (FFPE) samples from 1050 different in vitro and in vivo models of breast, prostate and lung cancers, as well as 364 clinical tumors from the same origin, were collected from 26 PREDECT collaborators across the EU. We established standard operating procedures for central processing of FFPE samples, including tissue microarray (TMA) construction and immunohistochemistry (IHC) with 15 different antibodies (CK8, Ki67, p-histone H3, ER, AR, p-AKT, p-ERK, p-p38, γH2AX, cleaved caspase 3, p-MET, HIF1α, p63, vimentin, E-cadherin). We constructed 50 TMA blocks, from which sections were cut and stained as well as digitized using whole slide imaging. Images were hosted on a WebMicroscope digital pathology platform and sample metadata on a PREDECT Metadata database (MBase). We developed image analysis methods for the detection and quantification of IHC biomarkers in the 48,800 stained TMA spots. As a proof-of-concept, we compared MCF-7 on several preclinical platforms including cell cultures, xenografts and xenograft tissue slices. Our results of the integrated biomarker phenotype suggest that of the various MCF-7 in vivo and ex vivo complex cell culture models, the xenograft tissue slice model was the most similar model platform to human clinical samples. In summary, we established a systems pathology approach to analyse and compare novel preclinical cancer models with IHC and digital imaging. The intention is that this large database will be made publicly available on the web as images and summary data that could be broadly useful to the community of cancer researchers and drug developers in comparing cancer model systems. The established infrastructure and workflow integrating molecular and digital pathology in a large-scale consortium setting could be applied to quantitative characterisation of consortium data in collaborations similar to PREDECT.
Citation Format: Sami Blom, Yinhai Wang, Tauno Metsalu, Tiina Vesterinen, Teijo Pellinen, Anne Grote, Nina Linder, Jenni Säilä, Katja Välimäki, Ruusu-Maria Kovanen, Outi Monni, Panu Kovanen, Emma Davies, Kristin Stock, Marta Estrada, Georgios Sflomos, Sylvia Grünewald, Catarina Brito, Julia Schüler, Ronald de Hoogt, Cathrin Brisken, Heiko van der Kuip, Wytske van Weerden, Simon Barry, Wolgang Sommergruber, Elizabeth Anderson, Matthias Nees, Juha Klefström, Jaak Vilo, Emmy Verschuren, Ralph Graeser, John Hickman, Johan Lundin, Olli Kallioniemi. Systems pathology for characterization of cancer model systems in a multicenter IMI-PREDECT project. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1698. doi:10.1158/1538-7445.AM2015-1698.
Rajaram, R.D.
Buric, D.
Caikovski, M.
Ayyanan, A.
Rougemont, J.
Shan, J.
Vainio, S.J.
Yalcin-Ozuysal, O.
Brisken, C.
(2015). Progesterone and Wnt4 control mammary stem cells via myoepithelial crosstalk. Embo j,
Vol.34
(5),
pp. 641-652.
show abstract
full text
Ovarian hormones increase breast cancer risk by poorly understood mechanisms. We assess the role of progesterone on global stem cell function by serially transplanting mouse mammary epithelia. Progesterone receptor (PR) deletion severely reduces the regeneration capacity of the mammary epithelium. The PR target, receptor activator of Nf-κB ligand (RANKL), is not required for this function, and the deletion of Wnt4 reduces the mammary regeneration capacity even more than PR ablation. A fluorescent reporter reveals so far undetected perinatal Wnt4 expression that is independent of hormone signaling. Pubertal and adult Wnt4 expression is specific to PR+ luminal cells and requires intact PR signaling. Conditional deletion of Wnt4 reveals that this early, previously unappreciated, Wnt4 expression is functionally important. We provide genetic evidence that canonical Wnt signaling in the myoepithelium required PR and Wnt4, whereas the canonical Wnt signaling activities observed in the embryonic mammary bud and in the stroma around terminal end buds are independent of Wnt4. Thus, progesterone and Wnt4 control stem cell function through a luminal-myoepithelial crosstalk with Wnt4 acting independent of PR perinatally..
Sflomos, G.
Shamsheddin, M.
Brisken, C.
(2015). An ex vivo model to study hormone action in the human breast. J vis exp,
(95),
p. e52436.
show abstract
full text
The study of hormone action in the human breast has been hampered by lack of adequate model systems. Upon in vitro culture, primary mammary epithelial cells tend to lose hormone receptor expression. Widely used hormone receptor positive breast cancer cell lines are of limited relevance to the in vivo situation. Here, we describe an ex vivo model to study hormone action in the human breast. Fresh human breast tissue specimens from surgical discard material such as reduction mammoplasties or mammectomies are mechanically and enzymatically digested to obtain tissue fragments containing ducts and lobules and multiple stromal cell types. These tissue microstructures kept in basal medium without growth factors preserve their intercellular contacts, the tissue architecture, and remain hormone responsive for several days. They are readily processed for RNA and protein extraction, histological analysis or stored in freezing medium. Fluorescence activated cell sorting (FACS) can be used to enrich for specific cell populations. This protocol provides a straightforward, standard approach for translational studies with highly complex, varied human specimens..
Procopio, M.-.
Laszlo, C.
Al Labban, D.
Kim, D.E.
Bordignon, P.
Jo, S.-.
Goruppi, S.
Menietti, E.
Ostano, P.
Ala, U.
Provero, P.
Hoetzenecker, W.
Neel, V.
Kilarski, W.W.
Swartz, M.A.
Brisken, C.
Lefort, K.
Dotto, G.P.
(2015). Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation. Nat cell biol,
Vol.17
(9),
pp. 1193-1204.
show abstract
full text
Stromal fibroblast senescence has been linked to ageing-associated cancer risk. However, density and proliferation of cancer-associated fibroblasts (CAFs) are frequently increased. Loss or downmodulation of the Notch effector CSL (also known as RBP-Jκ) in dermal fibroblasts is sufficient for CAF activation and ensuing keratinocyte-derived tumours. We report that CSL silencing induces senescence of primary fibroblasts from dermis, oral mucosa, breast and lung. CSL functions in these cells as a direct repressor of multiple senescence- and CAF-effector genes. It also physically interacts with p53, repressing its activity. CSL is downmodulated in stromal fibroblasts of premalignant skin actinic keratosis lesions and squamous cell carcinomas, whereas p53 expression and function are downmodulated only in the latter, with paracrine FGF signalling as the probable culprit. Concomitant loss of CSL and p53 overcomes fibroblast senescence, enhances expression of CAF effectors and promotes stromal and cancer cell expansion. The findings support a CAF activation-stromal co-evolution model under convergent CSL-p53 control..
Brisken, C.
Hess, K.
Jeitziner, R.
(2015). Progesterone and Overlooked Endocrine Pathways in Breast Cancer Pathogenesis. Endocrinology,
Vol.156
(10),
pp. 3442-3450.
show abstract
Worldwide, breast cancer incidence has been increasing for decades. Exposure to reproductive hormones, as occurs with recurrent menstrual cycles, affects breast cancer risk, and can promote disease progression. Exogenous hormones and endocrine disruptors have also been implicated in increasing breast cancer incidence. Numerous in vitro studies with hormone-receptor-positive cell lines have provided insights into the complexities of hormone receptor signaling at the molecular level; in vivo additional layers of complexity add on to this. The combined use of mouse genetics and tissue recombination techniques has made it possible to disentangle hormone action in vivo and revealed that estrogens, progesterone, and prolactin orchestrate distinct developmental stages of mammary gland development. The 2 ovarian steroids that fluctuate during menstrual cycles act on a subset of mammary epithelial cells, the hormone-receptor-positive sensor cells, which translate and amplify the incoming systemic signals into local, paracrine stimuli. Progesterone has emerged as a major regulator of cell proliferation and stem cell activation in the adult mammary gland. Two progesterone receptor targets, receptor activator of NfκB ligand and Wnt4, serve as downstream paracrine mediators of progesterone receptor-induced cell proliferation and stem cell activation, respectively. Some of the findings in the mouse have been validated in human ex vivo models and by next-generation whole-transcriptome sequencing on healthy donors staged for their menstrual cycles. The implications of these insights into the basic control mechanisms of mammary gland development for breast carcinogenesis and the possible role of endocrine disruptors, in particular bisphenol A in this context, will be discussed below..
Brisken, C.
Ataca, D.
(2015). Endocrine hormones and local signals during the development of the mouse mammary gland. Wiley interdiscip rev dev biol,
Vol.4
(3),
pp. 181-195.
show abstract
Most of mammary gland development occurs postnatally under the control of female reproductive hormones, which in turn interact with other endocrine factors. While hormones impinge on many tissues and trigger very complex biological responses, tissue recombination experiments with hormone receptor-deficient mammary epithelia revealed eminent roles for estrogens, progesterone, and prolactin receptor (PrlR) signaling that are intrinsic to the mammary epithelium. A subset of the luminal mammary epithelial cells expresses the estrogen receptor α (ERα), the progesterone receptor (PR), and the PrlR and act as sensor cells. These cells convert the detected systemic signals into local signals that are developmental stage-dependent and may be direct, juxtacrine, or paracrine. This setup ensures that the original input is amplified and that the biological responses of multiple cell types can be coordinated. Some key mediators of hormone action have been identified such as Wnt, EGFR, IGFR, and RANK signaling. Multiple signaling pathways such as FGF, Hedgehog, and Notch signaling participate in driving different aspects of mammary gland development locally but how they link to the hormonal control remains to be elucidated. An increasing number of endocrine factors are appearing to have a role in mammary gland development, the adipose tissue is increasingly recognized to play a role in endocrine regulation, and a complex role of the immune system with multiple different cell types is being revealed. For further resources related to this article, please visit the WIREs website..
Hickman, J.A.
Graeser, R.
de Hoogt, R.
Vidic, S.
Brito, C.
Gutekunst, M.
van der Kuip, H.
IMI PREDECT Consortium,
(2014). Three-dimensional models of cancer for pharmacology and cancer cell biology: capturing tumor complexity in vitro/ex vivo. Biotechnol j,
Vol.9
(9),
pp. 1115-1128.
show abstract
Cancers are complex and heterogeneous pathological "organs" in a dynamic interplay with their host. Models of human cancer in vitro, used in cancer biology and drug discovery, are generally highly reductionist. These cancer models do not incorporate complexity or heterogeneity. This raises the question as to whether the cancer models' biochemical circuitry (not their genome) represents, with sufficient fidelity, a tumor in situ. Around 95% of new anticancer drugs eventually fail in clinical trial, despite robust indications of activity in existing in vitro pre-clinical models. Innovative models are required that better capture tumor biology. An important feature of all tissues, and tumors, is that cells grow in three dimensions. Advances in generating and characterizing simple and complex (with added stromal components) three-dimensional in vitro models (3D models) are reviewed in this article. The application of stirred bioreactors to permit both scale-up/scale-down of these cancer models and, importantly, methods to permit controlled changes in environment (pH, nutrients, and oxygen) are also described. The challenges of generating thin tumor slices, their utility, and potential advantages and disadvantages are discussed. These in vitro/ex vivo models represent a distinct move to capture the realities of tumor biology in situ, but significant characterization work still remains to be done in order to show that their biochemical circuitry accurately reflects that of a tumor..
Cimino, D.
De Pittà, C.
Orso, F.
Zampini, M.
Casara, S.
Penna, E.
Quaglino, E.
Forni, M.
Damasco, C.
Pinatel, E.
Ponzone, R.
Romualdi, C.
Brisken, C.
De Bortoli, M.
Biglia, N.
Provero, P.
Lanfranchi, G.
Taverna, D.
(2013). miR148b is a major coordinator of breast cancer progression in a relapse-associated microRNA signature by targeting ITGA5, ROCK1, PIK3CA, NRAS, and CSF1. Faseb j,
Vol.27
(3),
pp. 1223-1235.
show abstract
Breast cancer is often fatal during its metastatic dissemination. To unravel the role of microRNAs (miRs) during malignancy, we analyzed miR expression in 77 primary breast carcinomas and identified 16 relapse-associated miRs that correlate with survival and/or distinguish tumor subtypes in different datasets. Among them, miR-148b, down-regulated in aggressive breast tumors, was found to be a major coordinator of malignancy. In fact, it is able to oppose various steps of tumor progression when overexpressed in cell lines by influencing invasion, survival to anoikis, extravasation, lung metastasis formation, and chemotherapy response. miR-148b controls malignancy by coordinating a novel pathway involving over 130 genes and, in particular, it directly targets players of the integrin signaling, such as ITGA5, ROCK1, PIK3CA/p110α, and NRAS, as well as CSF1, a growth factor for stroma cells. Our findings reveal the importance of the identified 16 miRs for disease outcome predictions and suggest a critical role for miR-148b in the control of breast cancer progression..
Brisken, C.
(2013). Progesterone signalling in breast cancer: a neglected hormone coming into the limelight. Nat rev cancer,
Vol.13
(6),
pp. 385-396.
show abstract
Understanding the biology of the breast and how ovarian hormones impinge on it is key to rational new approaches in breast cancer prevention and therapy. Because of the success of selective oestrogen receptor modulators (SERMs), such as tamoxifen, and aromatase inhibitors in breast cancer treatment, oestrogens have long received the most attention. Early progesterone receptor (PR) antagonists, however, were dismissed because of severe side effects, but awareness is now increasing that progesterone is an important hormone in breast cancer. Oestrogen receptor-α (ERα) signalling and PR signalling have distinct roles in normal mammary gland biology in mice; both ERα and PR delegate many of their biological functions to distinct paracrine mediators. If the findings in the mouse model translate to humans, new preventive and therapeutic perspectives might open up..
Soto, A.M.
Brisken, C.
Schaeberle, C.
Sonnenschein, C.
(2013). Does cancer start in the womb? altered mammary gland development and predisposition to breast cancer due to in utero exposure to endocrine disruptors. J mammary gland biol neoplasia,
Vol.18
(2),
pp. 199-208.
show abstract
full text
We are now witnessing a resurgence of theories of development and carcinogenesis in which the environment is again being accepted as a major player in phenotype determination. Perturbations in the fetal environment predispose an individual to disease that only becomes apparent in adulthood. For example, gestational exposure to diethylstilbestrol resulted in clear cell carcinoma of the vagina and breast cancer. In this review the effects of the endocrine disruptor bisphenol-A (BPA) on mammary development and tumorigenesis in rodents is used as a paradigmatic example of how altered prenatal mammary development may lead to breast cancer in humans who are also widely exposed to it through plastic goods, food and drink packaging, and thermal paper receipts. Changes in the stroma and its extracellular matrix led to altered ductal morphogenesis. Additionally, gestational and lactational exposure to BPA increased the sensitivity of rats and mice to mammotropic hormones during puberty and beyond, thus suggesting a plausible explanation for the increased incidence of breast cancer..
Tanos, T.
Sflomos, G.
Echeverria, P.C.
Ayyanan, A.
Gutierrez, M.
Delaloye, J.-.
Raffoul, W.
Fiche, M.
Dougall, W.
Schneider, P.
Yalcin-Ozuysal, O.
Brisken, C.
(2013). Progesterone/RANKL is a major regulatory axis in the human breast. Sci transl med,
Vol.5
(182),
p. 182ra55.
show abstract
Estrogens and progesterones are major drivers of breast development but also promote carcinogenesis in this organ. Yet, their respective roles and the mechanisms underlying their action in the human breast are unclear. Receptor activator of nuclear factor κB ligand (RANKL) has been identified as a pivotal paracrine mediator of progesterone function in mouse mammary gland development and mammary carcinogenesis. Whether the factor has the same role in humans is of clinical interest because an inhibitor for RANKL, denosumab, is already used for the treatment of bone disease and might benefit breast cancer patients. We show that progesterone receptor (PR) signaling failed to induce RANKL in PR(+) breast cancer cell lines and in dissociated, cultured breast epithelial cells. In clinical specimens from healthy donors and intact breast tissue microstructures, hormone response was maintained and RANKL expression was under progesterone control, which increased RNA stability. RANKL was sufficient to trigger cell proliferation and was required for progesterone-induced proliferation. The findings were validated in vivo where RANKL protein expression in the breast epithelium correlated with serum progesterone levels and the protein was expressed in a subset of luminal cells that express PR. Thus, important hormonal control mechanisms are conserved across species, making RANKL a potential target in breast cancer treatment and prevention..
Tanos, T.
Rojo, L.
Echeverria, P.
Brisken, C.
(2012). ER and PR signaling nodes during mammary gland development. Breast cancer res,
Vol.14
(4),
p. 210.
show abstract
The ovarian hormones estrogen and progesterone orchestrate postnatal mammary gland development and are implicated in breast cancer. Most of our understanding of the molecular mechanisms of estrogen receptor (ER) and progesterone receptor (PR) signaling stems from in vitro studies with hormone receptor-positive cell lines. They have shown that ER and PR regulate gene transcription either by binding to DNA response elements directly or via other transcription factors and recruiting co-regulators. In addition they cross-talk with other signaling pathways through nongenomic mechanisms. Mouse genetics combined with tissue recombination techniques have provided insights about the action of these two hormones in vivo. It has emerged that hormones act on a subset of mammary epithelial cells and relegate biological functions to paracrine factors. With regards to hormonal signaling in breast carcinomas, global gene expression analyses have led to the identification of gene expression signatures that are characteristic of ERα-positive tumors that have stipulated functional studies of hitherto poorly understood transcription factors. Here, we highlight what has been learned about ER and PR signaling nodes in these different systems and attempt to lay out in which way the insights may converge..
Rajaram, R.D.
Brisken, C.
(2012). Paracrine signaling by progesterone. Mol cell endocrinol,
Vol.357
(1-2),
pp. 80-90.
show abstract
Steroid hormones coordinate and control the development and function of many organs and are implicated in many pathological processes. Progesterone signaling, in particular, is essential for several important female reproductive functions. Physiological effects of progesterone are mediated by its cognate receptor, expressed in a subset of cells in target tissues. Experimental evidence has accumulated that progesterone acts through both cell intrinsic as well as paracrine signaling mechanisms. By relegating the hormonal stimulus to paracrine signaling cascades the systemic signal gets amplified locally and signaling reaches different cell types that are devoid of hormone receptors. Interestingly, distinct biological responses to progesterone in different target tissues rely on several tissue-specific and some common paracrine factors that coordinate biological responses in different cell types. Evidence is forthcoming that the intercellular signaling pathways that control development and physiological functions are important in tumorigenesis..
Ayyanan, A.
Laribi, O.
Schuepbach-Mallepell, S.
Schrick, C.
Gutierrez, M.
Tanos, T.
Lefebvre, G.
Rougemont, J.
Yalcin-Ozuysal, O.
Brisken, C.
(2011). Perinatal exposure to bisphenol a increases adult mammary gland progesterone response and cell number. Mol endocrinol,
Vol.25
(11),
pp. 1915-1923.
show abstract
Bisphenol A [BPA, 2,2,-bis (hydroxyphenyl) propane] is one of the highest-volume chemicals produced worldwide. It is detected in body fluids of more than 90% of the human population. Originally synthesized as an estrogenic compound, it is currently utilized to manufacture food and beverage containers resulting in uptake with food and drinks. There is concern that exposure to low doses of BPA, defined as less than or equal to 5 mg/kg body weight /d, may have developmental effects on various hormone-responsive organs including the mammary gland. Here, we asked whether perinatal exposure to a range of low doses of BPA is sufficient to alter mammary gland hormone response later on in life, with a possible impact on breast cancer risk. To mimic human exposure, we added BPA to the drinking water of C57/Bl6 breeding pairs. Analysis of the mammary glands of their daughters at puberty showed that estrogen-dependent transcriptional events were perturbed and the number of terminal end buds, estrogen-induced proliferative structures, was altered in a dose-dependent fashion. Importantly, adult females showed an increase in mammary epithelial cell numbers comparable to that seen in females exposed to diethylbestrol, a compound exposure to which was previously linked to increased breast cancer risk. Molecularly, the mRNAs encoding Wnt-4 and receptor activator of nuclear factor κB ligand, two key mediators of hormone function implicated in control of mammary stem cell proliferation and carcinogenesis, showed increased induction by progesterone in the mammary tissue of exposed mice. Thus, perinatal exposure to environmentally relevant doses of BPA alters long-term hormone response that may increase the propensity to develop breast cancer..
Dong, J.
Huang, S.
Caikovski, M.
Ji, S.
McGrath, A.
Custorio, M.G.
Creighton, C.J.
Maliakkal, P.
Bogoslovskaia, E.
Du, Z.
Zhang, X.
Lewis, M.T.
Sablitzky, F.
Brisken, C.
Li, Y.
(2011). ID4 regulates mammary gland development by suppressing p38MAPK activity. Development,
Vol.138
(23),
pp. 5247-5256.
show abstract
The ID family of helix-loop-helix proteins regulates cell proliferation and differentiation in many different developmental pathways, but the functions of ID4 in mammary development are unknown. We report that mouse Id4 is expressed in cap cells, basal cells and in a subset of luminal epithelial cells, and that its targeted deletion impairs ductal expansion and branching morphogenesis as well as cell proliferation induced by estrogen and/or progesterone. We discover that p38MAPK is activated in Id4-null mammary cells. p38MAPK is also activated following siRNA-mediated Id4 knockdown in transformed mammary cells. This p38MAPK activation is required for the reduced proliferation and increased apoptosis in Id4-ablated mammary glands. Therefore, ID4 promotes mammary gland development by suppressing p38MAPK activity..
Booth, B.W.
Boulanger, C.A.
Anderson, L.H.
Jimenez-Rojo, L.
Brisken, C.
Smith, G.H.
(2010). Amphiregulin mediates self-renewal in an immortal mammary epithelial cell line with stem cell characteristics. Exp cell res,
Vol.316
(3),
pp. 422-432.
show abstract
full text
Amphiregulin (AREG), a ligand for epidermal growth factor receptor, is required for mammary gland ductal morphogenesis and mediates estrogen actions in vivo, emerging as an essential growth factor during mammary gland growth and differentiation. The COMMA-D beta-geo (CDbetageo) mouse mammary cell line displays characteristics of normal mammary progenitor cells including the ability to regenerate a mammary gland when transplanted into the cleared fat pad of a juvenile mouse, nuclear label retention, and the capacity to form anchorage-independent mammospheres. We demonstrate that AREG is essential for formation of floating mammospheres by CDbetageo cells and that the mitogen activated protein kinase signaling pathway is involved in AREG-mediated mammosphere formation. Addition of exogenous AREG promotes mammosphere formation in cells where AREG expression is knocked down by siRNA and mammosphere formation by AREG(-/-) mammary epithelial cells. AREG knockdown inhibits mammosphere formation by duct-limited mammary progenitor cells but not lobule-limited mammary progenitor cells. These data demonstrate AREG mediates the function of a subset of mammary progenitor cells in vitro..
Dietrich, C.
Weiss, C.
Bockamp, E.
Brisken, C.
Roskams, T.
Morris, R.
Oesch-Bartlomowicz, B.
Oesch, F.
(2010). Stem cells in chemical carcinogenesis. Arch toxicol,
Vol.84
(3),
pp. 245-251.
Yang, C.
Chen, L.
Li, C.
Lynch, M.C.
Brisken, C.
Schmidt, E.V.
(2010). Cyclin D1 enhances the response to estrogen and progesterone by regulating progesterone receptor expression. Mol cell biol,
Vol.30
(12),
pp. 3111-3125.
show abstract
Estrogen and progesterone are the defining hormones of normal female development, and both play critical roles in breast carcinogenesis. Cyclin D1 is a breast cancer oncogene whose amplification is linked to poor prognosis in estrogen and progesterone receptor-positive breast cancers. Here we report that cyclin D1 regulates progesterone receptor expression, consequently enhancing responses to estrogen and progesterone. Estrogen treatment of cyclin D1 transgenic mice increased progesterone receptor expression and induced mammary hyperplasias that were stimulated by progesterone and blocked by a progesterone antagonist. Progesterone receptor levels decreased in cyclin D1 knockout mice. Cyclin D1 regulated progesterone receptor expression through a novel estrogen- and cyclin D1-responsive enhancer in DNA encoding part of the 3' untranslated region of the progesterone receptor gene. Small inhibitory RNAs for cyclin D1 decreased progesterone receptor expression and estrogen receptor binding to the 3' enhancer region in human breast cancer cells. Since estrogen and progesterone regulate cyclin D1, our results suggest that cyclin D1's participation in a feed-forward loop could contribute to increased breast cancer risks associated with estrogen and progesterone combinations. Additionally, its regulation of the progesterone receptor identifies a novel role for cyclin D1 in ovarian hormone control of breast development and breast carcinogenesis..
Battula, V.L.
Evans, K.W.
Hollier, B.G.
Shi, Y.
Marini, F.C.
Ayyanan, A.
Wang, R.-.
Brisken, C.
Guerra, R.
Andreeff, M.
Mani, S.A.
(2010). Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem cells,
Vol.28
(8),
pp. 1435-1445.
show abstract
The epithelial-to-mesenchymal transition (EMT) is an embryonic process that becomes latent in most normal adult tissues. Recently, we have shown that induction of EMT endows breast epithelial cells with stem cell traits. In this report, we have further characterized the EMT-derived cells and shown that these cells are similar to mesenchymal stem cells (MSCs) with the capacity to differentiate into multiple tissue lineages. For this purpose, we induced EMT by ectopic expression of Twist, Snail, or transforming growth factor-beta in immortalized human mammary epithelial cells. We found that the EMT-derived cells and MSCs share many properties including the antigenic profile typical of MSCs, that is, CD44(+), CD24(-), and CD45(-). Conversely, MSCs express EMT-associated genes, such as Twist, Snail, and mesenchyme forkhead 1 (FOXC2). Interestingly, CD140b (platelet-derived growth factor receptor-beta), a marker for naive MSCs, is exclusively expressed in EMT-derived cells and not in their epithelial counterparts. Moreover, functional analyses revealed that EMT-derived cells but not the control cells can differentiate into alizarin red S-positive mature osteoblasts, oil red O-positive adipocytes and alcian blue-positive chondrocytes similar to MSCs. We also observed that EMT-derived cells but not the control cells invade and migrate towards MDA-MB-231 breast cancer cells similar to MSCs. In vivo wound homing assays in nude mice revealed that the EMT-derived cells home to wound sites similar to MSCs. In conclusion, we have demonstrated that the EMT-derived cells are similar to MSCs in gene expression, multilineage differentiation, and ability to migrate towards tumor cells and wound sites..
Yalcin-Ozuysal, O.
Fiche, M.
Guitierrez, M.
Wagner, K.-.
Raffoul, W.
Brisken, C.
(2010). Antagonistic roles of Notch and p63 in controlling mammary epithelial cell fates. Cell death differ,
Vol.17
(10),
pp. 1600-1612.
show abstract
The breast epithelium has two major compartments, luminal and basal cells, that are established and maintained by poorly understood mechanisms. The p53 homolog, p63, is required for the formation of mammary buds, but its function in the breast after birth is unknown. We show that in primary human breast epithelial cells, maintenance of basal cell characteristics depends on continued expression of the p63 isoform, ΔNp63, which is expressed in the basal compartment. Forced expression of ΔNp63 in purified luminal cells confers a basal phenotype. Notch signaling downmodulates ΔNp63 expression and mimics ΔNp63 depletion, whereas forced expression of ΔNp63 partially counteracts the effects of Notch. Consistent with Notch activation specifying luminal cell fate in the mammary gland, Notch signaling activity is specifically detected in mice at sites of pubertal ductal morphogenesis where luminal cell fate is determined. Basal cells in which Notch signaling is active show decreased p63 expression. Both constitutive expression of ΔNp63 and ablation of Notch signaling are incompatible with luminal cell fate. Thus, the balance between basal and luminal cell compartments of the breast is regulated by antagonistic functions of ΔNp63 and Notch..
Brisken, C.
O'Malley, B.
(2010). Hormone action in the mammary gland. Cold spring harb perspect biol,
Vol.2
(12),
p. a003178.
show abstract
A woman's breast cancer risk is affected by her reproductive history. The hormonal milieu also influences the course of the disease. The female reproductive hormones, estrogens, progesterone, and prolactin, have a major impact on breast cancer and control postnatal mammary gland development. Analysis of hormone receptor mutant mouse strains combined with tissue recombination techniques and proteomics revealed that sequential activation of hormone signaling in the mammary epithelium is required for progression of morphogenesis. Hormones impinge on a subset of luminal mammary epithelial cells (MECs) that express hormone receptors and act as sensor cells translating and amplifying systemic signals into local stimuli. Proliferation is induced by paracrine mechanisms mediated by distinct factors at different stages. Tissue and stage specificity of hormonal signaling is achieved at the molecular level by different chromatin contexts and differential recruitment of coactivators and corepressors..
Hu, B.
Lefort, K.
Qiu, W.
Nguyen, B.-.
Rajaram, R.D.
Castillo, E.
He, F.
Chen, Y.
Angel, P.
Brisken, C.
Dotto, G.P.
(2010). Control of hair follicle cell fate by underlying mesenchyme through a CSL-Wnt5a-FoxN1 regulatory axis. Genes dev,
Vol.24
(14),
pp. 1519-1532.
show abstract
Epithelial-mesenchymal interactions are key to skin morphogenesis and homeostasis. We report that maintenance of the hair follicle keratinocyte cell fate is defective in mice with mesenchymal deletion of the CSL/RBP-Jkappa gene, the effector of "canonical" Notch signaling. Hair follicle reconstitution assays demonstrate that this can be attributed to an intrinsic defect of dermal papilla cells. Similar consequences on hair follicle differentiation result from deletion of Wnt5a, a specific dermal papilla signature gene that we found to be under direct Notch/CSL control in these cells. Functional rescue experiments establish Wnt5a as an essential downstream mediator of Notch-CSL signaling, impinging on expression in the keratinocyte compartment of FoxN1, a gene with a key hair follicle regulatory function. Thus, Notch/CSL signaling plays a unique function in control of hair follicle differentiation by the underlying mesenchyme, with Wnt5a signaling and FoxN1 as mediators..
Beleut, M.
Rajaram, R.D.
Caikovski, M.
Ayyanan, A.
Germano, D.
Choi, Y.
Schneider, P.
Brisken, C.
(2010). Two distinct mechanisms underlie progesterone-induced proliferation in the mammary gland. Proc natl acad sci u s a,
Vol.107
(7),
pp. 2989-2994.
show abstract
full text
The mouse mammary gland develops postnatally under the control of female reproductive hormones. Estrogens and progesterone trigger morphogenesis by poorly understood mechanisms acting on a subset of mammary epithelial cells (MECs) that express their cognate receptors, estrogen receptor alpha (ERalpha) and progesterone receptor (PR). Here, we show that in the adult female, progesterone drives proliferation of MECs in two waves. The first, small wave, encompasses PR(+) cells and requires cyclin D1, the second, large wave, comprises mostly PR(-) cells and relies on the tumor necrosis factor (TNF) family member, receptor activator of NF-kappaB-ligand (RANKL). RANKL elicits proliferation by a paracrine mechanism. Ablation of RANKL in the mammary epithelium blocks progesterone-induced morphogenesis, and ectopic expression of RANKL in MECs completely rescues the PR(-/-) phenotype. Systemic administration of RANKL triggers proliferation in the absence of PR signaling, and injection of a RANK signaling inhibitor interferes with progesterone-induced proliferation. Thus, progesterone elicits proliferation by a cell-intrinsic and a, more important, paracrine mechanism..
Cicalese, A.
Bonizzi, G.
Pasi, C.E.
Faretta, M.
Ronzoni, S.
Giulini, B.
Brisken, C.
Minucci, S.
Di Fiore, P.P.
Pelicci, P.G.
(2009). The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell,
Vol.138
(6),
pp. 1083-1095.
show abstract
Stem-like cells may be integral to the development and maintenance of human cancers. Direct proof is still lacking, mainly because of our poor understanding of the biological differences between normal and cancer stem cells (SCs). Using the ErbB2 transgenic model of breast cancer, we found that self-renewing divisions of cancer SCs are more frequent than their normal counterparts, unlimited and symmetric, thus contributing to increasing numbers of SCs in tumoral tissues. SCs with targeted mutation of the tumor suppressor p53 possess the same self-renewal properties as cancer SCs, and their number increases progressively in the p53 null premalignant mammary gland. Pharmacological reactivation of p53 correlates with restoration of asymmetric divisions in cancer SCs and tumor growth reduction, without significant effects on additional cancer cells. These data demonstrate that p53 regulates polarity of cell division in mammary SCs and suggest that loss of p53 favors symmetric divisions of cancer SCs, contributing to tumor growth..
Yalcin-Ozuysal, O.
Brisken, C.
(2009). From normal cell types to malignant phenotypes. Breast cancer res,
Vol.11
(6),
p. 306.
show abstract
The phenotypic diversity of breast cancer has been proposed to result from different target cell types undergoing oncogenic transformation and giving rise to cancer stem cells. Global gene expression profiling revealed distinct molecular phenotypes and some of these signatures were held to reflect the cell of origin, with the basal carcinomas arising from basal/progenitor cells. Recent work challenges this view by providing evidence that luminal precursor cells are involved in the pathogenesis of basal breast cancers and has made new links between normal cell populations and molecular tumor phenotypes..
Barbaroux, J.-.
Beleut, M.
Brisken, C.
Mueller, C.G.
Groves, R.W.
(2008). Epidermal receptor activator of NF-kappaB ligand controls Langerhans cells numbers and proliferation. J immunol,
Vol.181
(2),
pp. 1103-1108.
show abstract
Langerhans cells (LC) are the dendritic APC population of the epidermis, where they reside for long periods and are self-replicating. The molecular signals underlying these characteristics are unknown. The TNF superfamily member receptor activator of NF-kappaB ligand (RANKL, TNFSF11) has been shown to sustain viability of blood dendritic cells in addition to its role in promoting proliferation and differentiation of several cell types, notably osteoclasts. In this study, we have studied expression of the RANKL system in skin and have defined a key role for this molecule in LC homeostasis. In vitro and in vivo, human KC expressed RANKL and epidermal LC expressed cell surface RANK. In vitro, RANKL sustained CD34(+) progenitor-derived LC viability following 72-h cultures in cytokine-free medium (79.5 +/- 1% vs 55.2 +/- 5.7% live cells, respectively; n = 4; p < 0.05). In vivo, RANKL-deficient mice displayed a marked reduction in epidermal LC density (507.1 +/- 77.2 vs 873.6 +/- 41.6 LC per mm(2); n = 9; p < 0.05) and their proliferation was impaired without a detectable effect on apoptosis. These data indicate a key role for the RANKL system in the regulation of LC survival within the skin and suggest a regulatory role for KC in the maintenance of epidermal LC homeostasis..
Boulay, A.
Breuleux, M.
Stephan, C.
Fux, C.
Brisken, C.
Fiche, M.
Wartmann, M.
Stumm, M.
Lane, H.A.
Hynes, N.E.
(2008). The Ret receptor tyrosine kinase pathway functionally interacts with the ERalpha pathway in breast cancer. Cancer res,
Vol.68
(10),
pp. 3743-3751.
show abstract
A limited number of receptor tyrosine kinases (e.g., ErbB and fibroblast growth factor receptor families) have been genetically linked to breast cancer development. Here, we investigated the contribution of the Ret receptor tyrosine kinase to breast tumor biology. Ret was expressed in primary breast tumors and cell lines. In estrogen receptor (ER)alpha-positive MCF7 and T47D lines, the ligand (glial-derived neurotrophic factor) activated signaling pathways and increased anchorage-independent proliferation in a Ret-dependent manner, showing that Ret signaling is functional in breast tumor cells. Ret expression was induced by estrogens and Ret signaling enhanced estrogen-driven proliferation, highlighting the functional interaction of Ret and ER pathways. Furthermore, Ret was detected in primary cancers, and there were higher Ret levels in ERalpha-positive tumors. In summary, we showed that Ret is a novel proliferative pathway interacting with ER signaling in vitro. Expression of Ret in primary breast tumors suggests that Ret might be a novel therapeutic target in breast cancer..
Mani, S.A.
Guo, W.
Liao, M.-.
Eaton, E.N.
Ayyanan, A.
Zhou, A.Y.
Brooks, M.
Reinhard, F.
Zhang, C.C.
Shipitsin, M.
Campbell, L.L.
Polyak, K.
Brisken, C.
Yang, J.
Weinberg, R.A.
(2008). The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell,
Vol.133
(4),
pp. 704-715.
show abstract
The epithelial-mesenchymal transition (EMT) is a key developmental program that is often activated during cancer invasion and metastasis. We here report that the induction of an EMT in immortalized human mammary epithelial cells (HMLEs) results in the acquisition of mesenchymal traits and in the expression of stem-cell markers. Furthermore, we show that those cells have an increased ability to form mammospheres, a property associated with mammary epithelial stem cells. Independent of this, stem cell-like cells isolated from HMLE cultures form mammospheres and express markers similar to those of HMLEs that have undergone an EMT. Moreover, stem-like cells isolated either from mouse or human mammary glands or mammary carcinomas express EMT markers. Finally, transformed human mammary epithelial cells that have undergone an EMT form mammospheres, soft agar colonies, and tumors more efficiently. These findings illustrate a direct link between the EMT and the gain of epithelial stem cell properties..
Tanos, T.
Brisken, C.
(2008). What signals operate in the mammary niche?. Breast dis,
Vol.29,
pp. 69-82.
show abstract
Adult stem cells reside in a specialized microenvironment, the niche, which controls their behavior. As mammary stem cells, and consequently their niches, are still poorly defined, we look at better-characterized adult mammalian stem cell niches in the hematopoietic system and the skin. We attempt to define the mammary stem cell niche functionally, based on the widely used mammary fat pad reconstitution assay. We note that the concept of the niche needs to be extended from the specialized microenvironment described in the hematopoietic system, to a model that takes into account the macroenviroment, as recently shown in the skin, and systemic clues as we will illustrate for the mammary gland where the reproductive hormones are major determinants of stem cell activation. In fact, in the mammary gland a special type of stem cells is determined only during pregnancy. Reproductive hormones act on hormone receptor positive cells, sensor cells, in the mammary epithelium to induce paracrine signaling that leads to activation of stem cells. Some of the downstream mediators are in common with other niches such as Wnt and possibly Notch signaling. Other signals are specific to the mammary gland such as amphiregulin and RANKL..
Brisken, C.
Duss, S.
(2007). Stem cells and the stem cell niche in the breast: an integrated hormonal and developmental perspective. Stem cell rev,
Vol.3
(2),
pp. 147-156.
show abstract
The mammary gland is a unique organ in that it undergoes most of its development after birth under the control of systemic hormones. Whereas in most other organs stem cells divide in response to local stimuli, to replace lost cells, in the mammary gland large numbers of cells need to be generated at specific times during puberty, estrous cycles and pregnancy to generate new tissue structures. This puts special demands on the mammary stem cells and requires coordination of local events with systemic needs. Our aim is to understand how the female reproductive hormones control mammary gland development and influence tumorigenesis. We have shown that steroid hormones act in a paracrine fashion in the mammary gland delegating different functions to locally produced factors. These in turn, affect cell-cell interactions that result in changes of cell behavior required for morphogenesis and differentiation. Here, we discuss how these hormonally regulated paracrine interactions may impinge on stem cells and the stem cell niche and how this integration of signals adds extra levels of complexity to current mammary stem cell models. We propose a model whereby the stem cell niches change depending on the developmental stages and the hormonal milieu. According to this model, repeated hormone stimulation of stem cells and their niches in the course of menstrual cycles may be an important early event in breast carcinogenesis and may explain the conundrum why breast cancer risk increases with the number of menstrual cycles experienced prior to a first pregnancy..
Ciarloni, L.
Mallepell, S.
Brisken, C.
(2007). Amphiregulin is an essential mediator of estrogen receptor alpha function in mammary gland development. Proc natl acad sci u s a,
Vol.104
(13),
pp. 5455-5460.
show abstract
full text
Most mammary gland development occurs after birth under the control of systemic hormones. Estrogens induce mammary epithelial cell proliferation during puberty via epithelial estrogen receptor alpha (ERalpha) by a paracrine mechanism. Epidermal growth factor receptor (EGFR) signaling has long been implicated downstream of ERalpha signaling, and several EGFR ligands have been described as estrogen-target genes in tumor cell lines. Here, we show that amphiregulin is the unique EGF family member to be transcriptionally induced by estrogen in the mammary glands of puberal mice at a time of exponential expansion of the ductal system. In fact, we find that estrogens induce amphiregulin through the ERalpha and require amphiregulin to induce proliferation of the mammary epithelium. Like ERalpha, amphiregulin is required in the epithelium of puberal mice for epithelial proliferation, terminal end buds formation, and ductal elongation. Subsequent stages, such as side-branching and alveologenesis, are not affected. When amphiregulin(-/-) mammary epithelial cells are in close vicinity to wild-type cells, they proliferate and contribute to all cell compartments of the ductal outgrowth. Thus, amphiregulin is an important paracrine mediator of estrogen function specifically required for puberty-induced ductal elongation, but not for any earlier or later developmental stages..
Duss, S.
André, S.
Nicoulaz, A.-.
Fiche, M.
Bonnefoi, H.
Brisken, C.
Iggo, R.D.
(2007). An oestrogen-dependent model of breast cancer created by transformation of normal human mammary epithelial cells. Breast cancer res,
Vol.9
(3),
p. R38.
show abstract
INTRODUCTION: About 70% of breast cancers express oestrogen receptor alpha (ESR1/ERalpha) and are oestrogen-dependent for growth. In contrast with the highly proliferative nature of ERalpha-positive tumour cells, ERalpha-positive cells in normal breast tissue rarely proliferate. Because ERalpha expression is rapidly lost when normal human mammary epithelial cells (HMECs) are grown in vitro, breast cancer models derived from HMECs are ERalpha-negative. Currently only tumour cell lines are available to model ERalpha-positive disease. To create an ERalpha-positive breast cancer model, we have forced normal HMECs derived from reduction mammoplasty tissue to express ERalpha in combination with other relevant breast cancer genes. METHODS: Candidate genes were selected based on breast cancer microarray data and cloned into lentiviral vectors. Primary HMECs prepared from reduction mammoplasty tissue were infected with lentiviral particles. Infected HMECs were characterised by Western blotting, immunofluorescence microscopy, microarray analysis, growth curves, karyotyping and SNP chip analysis. The tumorigenicity of the modified HMECs was tested after orthotopic injection into the inguinal mammary glands of NOD/SCID mice. Cells were marked with a fluorescent protein to allow visualisation in the fat pad. The growth of the graft was analysed by fluorescence microscopy of the mammary glands and pathological analysis of stained tissue sections. Oestrogen dependence of tumour growth was assessed by treatment with the oestrogen antagonist fulvestrant. RESULTS: Microarray analysis of ERalpha-positive tumours reveals that they commonly overexpress the Polycomb-group gene BMI1. Lentiviral transduction with ERalpha, BMI1, TERT and MYC allows primary HMECs to be expanded in vitro in an oestrogen-dependent manner. Orthotopic xenografting of these cells into the mammary glands of NOD/SCID mice results in the formation of ERalpha-positive tumours that metastasise to multiple organs. The cells remain wild type for TP53, diploid and genetically stable. In vivo tumour growth and in vitro proliferation of cells explanted from tumours are dependent on oestrogen. CONCLUSION: We have created a genetically defined model of ERalpha-positive human breast cancer based on normal HMECs that has the potential to model human oestrogen-dependent breast cancer in a mouse and enables the study of mechanisms involved in tumorigenesis and metastasis..
Ayyanan, A.
Civenni, G.
Ciarloni, L.
Morel, C.
Mueller, N.
Lefort, K.
Mandinova, A.
Raffoul, W.
Fiche, M.
Dotto, G.P.
Brisken, C.
(2006). Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proc natl acad sci u s a,
Vol.103
(10),
pp. 3799-3804.
show abstract
full text
Wnt and Notch signaling have long been established as strongly oncogenic in the mouse mammary gland. Aberrant expression of several Wnts and other components of this pathway in human breast carcinomas has been reported, but evidence for a causative role in the human disease has been missing. Here we report that increased Wnt signaling, as achieved by ectopic expression of Wnt-1, triggers the DNA damage response (DDR) and an ensuing cascade of events resulting in tumorigenic conversion of primary human mammary epithelial cells. Wnt-1-transformed cells have high telomerase activity and compromised p53 and Rb function, grow as spheres in suspension, and in mice form tumors that closely resemble medullary carcinomas of the breast. Notch signaling is up-regulated through a mechanism involving increased expression of the Notch ligands Dll1, Dll3, and Dll4 and is required for expression of the tumorigenic phenotype. Increased Notch signaling in primary human mammary epithelial cells is sufficient to reproduce some aspects of Wnt-induced transformation. The relevance of these findings for human breast cancer is supported by the fact that expression of Wnt-1 and Wnt-4 and of established Wnt target genes, such as Axin-2 and Lef-1, as well as the Notch ligands, such as Dll3 and Dll4, is up-regulated in human breast carcinomas..
Brisken, C.
Rajaram, R.D.
(2006). Alveolar and lactogenic differentiation. J mammary gland biol neoplasia,
Vol.11
(3-4),
pp. 239-248.
show abstract
The mouse mammary gland is a complex tissue that proliferates and differentiates under the control of systemic hormones during puberty, pregnancy and lactation. Once a highly branched milk duct system has been established, during mid/late pregnancy, alveoli, little saccular outpouchings, sprout all over the ductal system and differentiate to become the sites of milk secretion. Here, we review the emerging network of the signaling pathways that connects hormonal stimuli with locally produced signaling molecules and the components of intracellular pathways that regulate alveologenesis and lactation. The powerful tools of mouse genetics have been instrumental in uncovering many of the signaling components involved in controlling alveolar and lactogenic differentiation..
Mallepell, S.
Krust, A.
Chambon, P.
Brisken, C.
(2006). Paracrine signaling through the epithelial estrogen receptor alpha is required for proliferation and morphogenesis in the mammary gland. Proc natl acad sci u s a,
Vol.103
(7),
pp. 2196-2201.
show abstract
Estradiol is a major regulator of postnatal mammary gland development and thought to exert its effects through estrogen receptor alpha (ERalpha) expressed in the mammary gland stroma and epithelium. Previous studies, however, were confounded by the use of an ERalpha mutant strain that retains some of the protein with transactivation activity. Here, we use an ERalpha-/- mouse strain in which no ERalpha transcript can be detected to analyze mammary gland development in the complete absence of ERalpha signaling. The ERalpha-/- females show no development beyond a rudimentary ductal system. By grafting ERalpha-/- epithelium or stroma in combination with ERalpha WT stroma or epithelium, we show that the primary target for estradiol is the mammary epithelium, whereas a direct response of the mammary stroma is not required for mammary gland development to proceed normally. Mammary glands reconstituted with ERalpha-/- mammary epithelium exposed to pregnancy hormones show increased transcription of milk protein genes, indicating that ERalpha signaling is not an absolute requirement for a transcriptional response to pregnancy hormones. When ERalpha-/- mammary epithelial cells are in close vicinity to ERalpha WT cells, they proliferate and contribute to all aspects of mammary gland development, indicating that estradiol, like progesterone, orchestrates proliferation and morphogenesis by a paracrine mechanism, affecting nearby cells in the mammary epithelium..
Farmer, P.
Bonnefoi, H.
Becette, V.
Tubiana-Hulin, M.
Fumoleau, P.
Larsimont, D.
Macgrogan, G.
Bergh, J.
Cameron, D.
Goldstein, D.
Duss, S.
Nicoulaz, A.-.
Brisken, C.
Fiche, M.
Delorenzi, M.
Iggo, R.
(2005). Identification of molecular apocrine breast tumours by microarray analysis. Oncogene,
Vol.24
(29),
pp. 4660-4671.
show abstract
Previous microarray studies on breast cancer identified multiple tumour classes, of which the most prominent, named luminal and basal, differ in expression of the oestrogen receptor alpha gene (ER). We report here the identification of a group of breast tumours with increased androgen signalling and a 'molecular apocrine' gene expression profile. Tumour samples from 49 patients with large operable or locally advanced breast cancers were tested on Affymetrix U133A gene expression microarrays. Principal components analysis and hierarchical clustering split the tumours into three groups: basal, luminal and a group we call molecular apocrine. All of the molecular apocrine tumours have strong apocrine features on histological examination (P=0.0002). The molecular apocrine group is androgen receptor (AR) positive and contains all of the ER-negative tumours outside the basal group. Kolmogorov-Smirnov testing indicates that oestrogen signalling is most active in the luminal group, and androgen signalling is most active in the molecular apocrine group. ERBB2 amplification is commoner in the molecular apocrine than the other groups. Genes that best split the three groups were identified by Wilcoxon test. Correlation of the average expression profile of these genes in our data with the expression profile of individual tumours in four published breast cancer studies suggest that molecular apocrine tumours represent 8-14% of tumours in these studies. Our data show that it is possible with microarray data to divide mammary tumour cells into three groups based on steroid receptor activity: luminal (ER+ AR+), basal (ER- AR-) and molecular apocrine (ER- AR+)..
Devgan, V.
Mammucari, C.
Millar, S.E.
Brisken, C.
Dotto, G.P.
(2005). p21WAF1/Cip1 is a negative transcriptional regulator of Wnt4 expression downstream of Notch1 activation. Genes dev,
Vol.19
(12),
pp. 1485-1495.
show abstract
In keratinocytes, the cyclin/CDK inhibitor p21(WAF1/Cip1) is a direct transcriptional target of Notch1 activation; loss of either the p21 or Notch1 genes expands stem cell populations and facilitates tumor development. The Notch1 tumor-suppressor function was associated with down-regulation of Wnt signaling. Here, we show that suppression of Wnt signaling by Notch1 activation is mediated, at least in part, by down-modulation of Wnts gene expression. p21 is a negative regulator of Wnts transcription downstream of Notch1 activation, independently of effects on the cell cycle. More specifically, expression of the Wnt4 gene is under negative control of endogenous p21 both in vitro and in vivo. p21 associates with the E2F-1 transcription factor at the Wnt4 promoter and causes curtailed recruitment of c-Myc and p300, and histone hypoacetylation at this promoter. Thus, p21 acts as a selective negative regulator of transcription and links the Notch and Wnt signaling pathways in keratinocyte growth control..
Chu, E.Y.
Hens, J.
Andl, T.
Kairo, A.
Yamaguchi, T.P.
Brisken, C.
Glick, A.
Wysolmerski, J.J.
Millar, S.E.
(2004). Canonical WNT signaling promotes mammary placode development and is essential for initiation of mammary gland morphogenesis. Development,
Vol.131
(19),
pp. 4819-4829.
show abstract
Mammary glands, like other skin appendages such as hair follicles and teeth, develop from the surface epithelium and underlying mesenchyme; however, the molecular controls of embryonic mammary development are largely unknown. We find that activation of the canonical WNT/beta-catenin signaling pathway in the embryonic mouse mammary region coincides with initiation of mammary morphogenesis, and that WNT pathway activity subsequently localizes to mammary placodes and buds. Several Wnt genes are broadly expressed in the surface epithelium at the time of mammary initiation, and expression of additional Wnt and WNT pathway genes localizes to the mammary lines and placodes as they develop. Embryos cultured in medium containing WNT3A or the WNT pathway activator lithium chloride (LiCl) display accelerated formation of expanded placodes, and LiCl induces the formation of ectopic placode-like structures that show elevated expression of the placode marker Wnt10b. Conversely, expression of the secreted WNT inhibitor Dickkopf 1 in transgenic embryo surface epithelium in vivo completely blocks mammary placode formation and prevents localized expression of all mammary placode markers tested. These data indicate that WNT signaling promotes placode development and is required for initiation of mammary gland morphogenesis. WNT signals play similar roles in hair follicle formation and thus may be broadly required for induction of skin appendage morphogenesis..
Brisken, C.
Ayyannan, A.
Nguyen, C.
Heineman, A.
Reinhardt, F.
Tan, J.
Dey, S.K.
Dotto, G.P.
Weinberg, R.A.
(2003). IGF-2 Is a Mediator of Prolactin-Induced Morphogenesis in the Breast. Developmental cell,
Vol.4
(2),
pp. 283-283.
Gass, S.
Harris, J.
Ormandy, C.
Brisken, C.
(2003). Using gene expression arrays to elucidate transcriptional profiles underlying prolactin function. J mammary gland biol neoplasia,
Vol.8
(3),
pp. 269-285.
show abstract
Prolactin is an ancient hormone, with different functions in many species. The binding of prolactin to its receptor, a member of the cytokine receptor superfamily, results in the activation of different intracellular signaling pathways, such as JAK2/STAT5, MAP kinase, and PI3K/AKT. How prolactin elicits so many different biological responses remains unclear. Recently, microarray technology has been applied to identify prolactin target genes in different systems. Here, we attempt to summarize and compare the available data. Our comparison of the genes reported to be transcriptionally regulated by prolactin indicates that there are few genes in common between the different tissues. Among the organs studied, mammary and prostate glands displayed the largest number of overlaps in putative prolactin target genes. Some of the candidates have been implicated in tumorigenesis. The relevance and validation of microarray data, as well as comparison of the results obtained by different groups, will be discussed..
Brisken, C.
(2002). Hormonal control of alveolar development and its implications for breast carcinogenesis. J mammary gland biol neoplasia,
Vol.7
(1),
pp. 39-48.
show abstract
During puberty and pregnancy, the breast undergoes major restructuring in order to produce a structure that can secrete and eject copious amounts of milk. By analogy to other branched organs such as the lung or the salivary gland, a large increase in surface area of the specialized epithelium is achieved through repeated ramifications of a system of ducts and alveoli arising from the nipple. In the breast, this process culminates in the appearance of thousands of alveoli or acini, saccular outpouchings from the ductal system. This paper focuses on this final stage of proliferation, the formation of alveolar structures and its control by systemic hormones..
Brisken, C.
Ayyannan, A.
Nguyen, C.
Heineman, A.
Reinhardt, F.
Tan, J.
Dey, S.K.
Dotto, G.P.
Weinberg, R.A.
(2002). IGF-2 is a mediator of prolactin-induced morphogenesis in the breast. Dev cell,
Vol.3
(6),
pp. 877-887.
show abstract
The mechanisms by which prolactin controls proliferation of mammary epithelial cells (MECs) and morphogenesis of the breast epithelium are poorly understood. We show that cyclin D1(-/-) MECs fail to proliferate in response to prolactin and identify IGF-2 as a downstream target of prolactin signaling that lies upstream of cyclin D1 transcription. Ectopic IGF-2 expression restores alveologenesis in prolactin receptor(-/-) epithelium. Alveologenesis is retarded in IGF-2-deficient MECs. IGF-2 and prolactin receptor mRNAs colocalize in the mammary epithelium. Prolactin induces IGF-2 mRNA and IGF-2 induces cyclin D1 protein in primary MECs. Thus, IGF-2 is a mediator of prolactin-induced alveologenesis; prolactin, IGF-2, and cyclin D1, all of which are overexpressed in breast cancers, are components of a developmental pathway in the mammary gland..
Brisken, C.
Socolovsky, M.
Lodish, H.F.
Weinberg, R.
(2002). The signaling domain of the erythropoietin receptor rescues prolactin receptor-mutant mammary epithelium. Proc natl acad sci u s a,
Vol.99
(22),
pp. 14241-14245.
show abstract
full text
The cytokine hormones prolactin and erythropoietin mediate tissue-specific developmental outcomes by activating their cognate receptors, prolactin receptor (PrlR) and erythropoietin receptor (EpoR), respectively. The EpoR is essential for red blood cell formation, whereas a principal function of PrlR is in the development of the mammary gland during pregnancy and lactation [Ormandy, C., et al. (1997) Genes Dev. 11, 167-178]. The instructive model of differentiation proposes that such distinct, cytokine-dependent developmental outcomes are a result of cytokine receptor-unique signals that bring about induction of lineage-specific genes. This view was challenged by our finding that an exogenously expressed PrlR could rescue EpoR(-/-) erythroid progenitors and mediate their differentiation into red blood cells. Together with similar findings in other hematopoietic lineages, this suggested that cytokine receptors do not play an instructive role in hematopoietic differentiation. Here, we show that these findings are not limited to the hematopoietic system but are of more general relevance to cytokine-dependent differentiation. We demonstrate that the developmental defect of PrlR(-/-) mammary epithelium is rescued by an exogenously expressed chimeric receptor (prl-EpoR) containing the PrlR extracellular domain joined to the EpoR transmembrane and intracellular domains. Like the wild-type PrlR, the prl-EpoR rescued alveologenesis and milk secretion in PrlR(-/-) mammary epithelium. These results suggest that, in cell types as unrelated as erythrocytes and mammary epithelial cells, cytokine receptors employ similar, generic signals that permit the expression of predetermined, tissue-specific differentiation programs..
Brisken, C.
Ayyanan, A.
Doppler, W.
(2002). Prolactin signaling and Stat5: going their own separate ways?. Breast cancer res,
Vol.4
(6),
pp. 209-212.
show abstract
Miyoshi et al. compared the role of the prolactin receptor (PrlR) and its downstream mediator, the signal transducer and activator of transcription 5 (Stat5), in mammary epithelial cells in vivo by studying PrlR-/- and Stat5ab-/- mouse mammary epithelial transplants during pregnancy. At first glance, the two mutant epithelia appear to have similar defects in the differentiation of the alveolar epithelium. However, a closer examination by Miyoshi et al. revealed defects in the epithelial architecture of the smallest ducts of Stat5ab-/- transplants not apparent in the PrlR-/- transplants, suggesting that Stat5 is more than a simple mediator of PrlR action..
Brisken, C.
Heineman, A.
Chavarria, T.
Elenbaas, B.
Tan, J.
Dey, S.K.
McMahon, J.A.
McMahon, A.P.
Weinberg, R.A.
(2000). Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling. Genes dev,
Vol.14
(6),
pp. 650-654.
show abstract
Female reproductive hormones control mammary gland morphogenesis. In the absence of the progesterone receptor (PR) from the mammary epithelium, ductal side-branching fails to occur. We can overcome this defect by ectopic expression of the protooncogene Wnt-1. Transplantation of mammary epithelia from Wnt-4(-)/(-) mice shows that Wnt-4 has an essential role in side-branching early in pregnancy. PR and Wnt-4 mRNAs colocalize to the luminal compartment of the ductal epithelium. Progesterone induces Wnt-4 in mammary epithelial cells and is required for increased Wnt-4 expression during pregnancy. Thus, Wnt signaling is essential in mediating progesterone function during mammary gland morphogenesis..
Brisken, C.
Kaur, S.
Chavarria, T.E.
Binart, N.
Sutherland, R.L.
Weinberg, R.A.
Kelly, P.A.
Ormandy, C.J.
(1999). Prolactin controls mammary gland development via direct and indirect mechanisms. Dev biol,
Vol.210
(1),
pp. 96-106.
show abstract
The inactivation of the prolactin receptor gene by homologous recombination has made it possible to investigate the role of prolactin signaling in mammary gland development without resort to ablative surgery of the endocrine glands. In knockout mice lacking the prolactin receptor, mammary development is normal up to puberty. Subsequently, the ducts branch less frequently than those of wild-type animals. While terminal end buds differentiate to alveolar buds in wild-type females by the end of puberty, in knockout females terminal end bud-like structures persist at the ductal ends. To distinguish between the developmental defects that are intrinsic to the epithelium and those that result from systemic endocrine alterations in prolactin receptor knockout mice, mammary epithelium from prolactin receptor knockouts was transplanted into mammary fat pads of wild-type mice. In virgin mice, the knockout epithelial transplants developed normally at puberty, indicating an indirect effect of prolactin on ductal development. Prolactin receptor knockout females are infertile due to multiple reproductive defects, but epithelial transplants allowed us to assess the extent to which the absence of prolactin receptor is limiting, under systemic conditions that allow full mammary gland development. During pregnancy, the prolactin receptor knockout transplants showed normal side branching and the formation of alveolar buds, but no lobuloalveolar development. Thus, prolactin affects mammary morphogenesis in two different ways: it controls ductal side branching and terminal end bud regression in virgin animals via indirect mechanisms, but acts directly on the mammary epithelium to produce lobuloalveolar development during pregnancy..
Brisken, C.
Park, S.
Vass, T.
Lydon, J.P.
O'Malley, B.W.
Weinberg, R.A.
(1998). A paracrine role for the epithelial progesterone receptor in mammary gland development. Proc natl acad sci u s a,
Vol.95
(9),
pp. 5076-5081.
show abstract
Recently generated progesterone receptor (PR)-negative (PR-/-) mice provide an excellent model for dissecting the role of progesterone in the development of the mammary gland during puberty and pregnancy. However, the full extent of the mammary gland defect in these mice caused by the absence of the PR cannot be assessed, because PR-/- mice do not exhibit estrous cycles and fail to become pregnant. To circumvent this difficulty, we have transplanted PR-/- breasts into wild-type mice, and we have demonstrated that the development of the mammary gland in the absence of the PR is arrested at the stage of the simple ductal system found in the young virgin mouse. Mammary transplants lacking the PR in the stromal compartment give rise to normal alveolar growth, whereas transplants containing PR-/- epithelium conserve the abnormal phenotype. Chimeric epithelia in which PR-/- cells are in close vicinity to PR wild-type cells go through complete alveolar development to which the PR-/- cells contribute. Together, these results indicate that progesterone acts by a paracrine mechanism on a subset of mammary epithelial cells to allow for alveolar growth and that expression of the PR is not required in all the cells of the mammary epithelium in order for alveolar development to proceed normally..
Brisken, C.
Endocrine Disruptors and Breast Cancer. Chimia,
Vol.62
(5),
pp. 406-406.
show abstract
Breast cancer strikes one out of eight women in Switzerland. The increase in breast cancer incidence over the past 70 years parallels an enormous increase of man-made, persistent chemicals in our environment; some of which have endocrine-disrupting properties in wildlife and/or in animal
models. Epidemiological evidence is strong that a woman's risk to get breast cancer is linked to her reproductive history and with that to the changes in her hormonal milieu. Exogenous hormones have also been shown to increase breast cancer risk, however, a causative link between exposure
to endocrine disruptors and human disease is difficult establish as many of these compounds are ubiquitous and no unexposed controls exist. The synthetic estrogen, diethylstilbestrol (DES), that was given to pregnant women for three decades, was banned because it was linked to a vaginal carcinoma
in their daughters. It has now been shown that not only women who have taken the drug themselves have increased breast cancer risk but also their daughters who were exposed in utero. This indicates that breast cancer risk can be affected by endocrine disruption not only in the adult
but already in utero. Evidence from animal models is accumulating that perinatal exposure to environmentally relevant, low doses of a related compound, bisphenol A (BPA), alters breast development and increases breast cancer risk. Given the prevalence of endocrine-disrupting agents
they deserve our attention.
.
Barbier, M.
Jaensch, S.
Cornelissen, F.
Vidic, S.
Gjerde, K.
de Hoogt, R.
Graeser, R.
Gustin, E.
Chong, Y.T.
Ellipsoid Segmentation Model for Analyzing Light-Attenuated 3D Confocal Image Stacks of Fluorescent Multi-Cellular Spheroids. Plos one,
Vol.11
(6),
pp. e0156942-e0156942.